1
|
Løkhammer S, Tesfaye M, Cabrera-Mendoza B, Sandås K, Pathak GA, Friligkou E, Le Hellard S, Polimanti R. Integration of Metabolomic and Brain Imaging Data Highlights Pleiotropy Among Posttraumatic Stress Disorder, Glycoprotein Acetyls, and Pallidum Structure. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2025; 5:100482. [PMID: 40270839 PMCID: PMC12013147 DOI: 10.1016/j.bpsgos.2025.100482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/16/2025] [Accepted: 03/01/2025] [Indexed: 04/25/2025] Open
Abstract
Background The development of posttraumatic stress disorder (PTSD) is attributable to the interplay between exposure to severe traumatic events, environmental factors, and biological characteristics. Blood and brain imaging markers have been associated with PTSD. However, to our knowledge, no study has systematically investigated the genetic relationship between PTSD, metabolic biomarkers, and brainwide imaging. Methods We integrated genome-wide data informative of PTSD, 233 metabolic biomarkers, and 3935 brain imaging-derived phenotypes (IDPs). Pleiotropy was assessed by applying global and local genetic correlation, colocalization, and genetically inferred causality. Results We observed significant genetic overlap between PTSD and glycoprotein acetyls (GlycA) (a stable inflammatory biomarker) in 2 independent cohorts (discovery r g = 0.26, p = 1.00 × 10-4; replication r g = 0.23, p = 5.99 × 10-19). Interestingly, there was no genetic correlation between anxiety and GlycA (p = .33). PTSD and GlycA were both genetically correlated with median T2∗ in the left pallidum (IDP-1444: r g = 0.14, p = 1.39 × 10-5; r g = -0.38, p = 2.50 × 10-3, respectively). Local genetic correlation between PTSD and GlycA was observed in 7 genetic regions (p < 2.0 × 10-5), mapping genes related to immune and stress response, inflammation, and metabolic processes. Furthermore, we identified 1 variant, rs12048743, with evidence of horizontal pleiotropy linking GlycA and IDP-1444 (z IDP-1444 = 17.14, z GlycA = -6.07, theta p = 2.06 × 10-8). Regional colocalization was observed among GlycA, IDP-1444, and tissue-specific transcriptomic regulation for brain frontal cortex and testis (rs12048743-chr1q32.1; posterior probability > 0.8). While we also tested causality between PTSD, metabolomic biomarkers, and brain IDPs, these were not consistent across different genetically informed causal inference methods. Conclusions Our findings highlight a new putative pleiotropic mechanism that links systemic inflammation and pallidum structure to PTSD.
Collapse
Affiliation(s)
- Solveig Løkhammer
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Dr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
| | - Markos Tesfaye
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Center for Precision Psychiatry, Division of Mental Health and Addiction, Oslo University Hospital & Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Brenda Cabrera-Mendoza
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut
- Veterans Affairs Connecticut Healthcare Center, West Haven, Connecticut
| | - Kristoffer Sandås
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Dr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
- School of Bioscience, University of Skövde, Skövde, Sweden
| | - Gita A. Pathak
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut
- Veterans Affairs Connecticut Healthcare Center, West Haven, Connecticut
| | - Eleni Friligkou
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut
- Veterans Affairs Connecticut Healthcare Center, West Haven, Connecticut
| | - Stéphanie Le Hellard
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Dr. Einar Martens Research Group for Biological Psychiatry, Center for Medical Genetics and Molecular Medicine, Haukeland University Hospital, Bergen, Norway
- Bergen Center for Brain Plasticity, Haukeland University Hospital, Bergen, Norway
| | - Renato Polimanti
- Department of Psychiatry, Yale School of Medicine, New Haven, Connecticut
- Veterans Affairs Connecticut Healthcare Center, West Haven, Connecticut
- Department of Chronic Disease Epidemiology, Yale School of Public Health, New Haven, Connecticut
- Department of Biomedical Informatics and Data Science, Yale School of Medicine, New Haven, Connecticut
- Wu Tsai Institute, Yale University, New Haven, Connecticut
| |
Collapse
|
2
|
He M, Zhang H, Luo Z, Duan X, Zhao F, Su P, Zeng Z, Zhou L, Chen C, Qiu J. Causal link between gut microbiota and obsessive-compulsive disorder: A two-sample Mendelian randomization analysis. J Affect Disord 2025; 379:852-860. [PMID: 40056996 DOI: 10.1016/j.jad.2025.02.099] [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: 07/27/2024] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND Previous studies have indicated a potential link between the gut microbiota and obsessive-compulsive disorder (OCD). However, the exact causal relationship remains uncertain. In this study, we employed a two-sample Mendelian randomization (MR) analysis to evaluate the causal connection between gut microbiota and OCD. METHODS We collected Genome-Wide Association Study (GWAS) summary data on gut microbiota (n = 18, 340) and OCD (n = 199, 169), using single nucleotide polymorphisms (SNPs) as the instrumental variable. SNPs with an F-statistic of <10 were deemed weak instrumental variables and subsequently excluded. The MR analysis was conducted using five methods: inverse variance weighting (IVW), MR Egger, weighted median, weighted mode, and simple mode. Heterogeneity and pleiotropy were assessed using Cochran's Q-test and MR Egger intercept test, while sensitivity analysis was performed using a leave-one-out approach. RESULTS The IVW analysis revealed that at the phylum level, Proteobacteria (OR = 0.545, 95%CI: 0.347-0.855, P = 0.008) served as a protective factor for OCD, whereas at the order level, Bacillales (OR = 1.327, 95%CI: 1.032-1.707, P = 0.027) was identified as a risk factor. At the family level, Ruminococcaceae (OR = 0.570, 95%CI: 0.354-0.918, P = 0.021) also acted as a protective factor. At the genus level, Bilophila (OR = 0.623, 95%CI: 0.425-0.911, P = 0.015) was a protective factor, while Eubacterium ruminantium group (OR = 1.347, 95%CI: 1.012-1.794, P = 0.041) and Lachnospiraceae UCG001 (OR = 1.384, 95%CI: 1.003-1.910, P = 0.048) were identified risk factors. Reverse MR analysis showed no significant causal relationship between OCD and the gut microbiota, with no significant heterogeneity or horizontal pleiotropy observed. CONCLUSION Our analysis suggested that specific gut microbiota might have a causal relationship with OCD, revealing potential intervention strategies for the prevention and treatment of this disorder.
Collapse
Affiliation(s)
- Mingjie He
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Hongyang Zhang
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Zheng Luo
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Xinhao Duan
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Feng Zhao
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Peng Su
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Zhijun Zeng
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Lixiao Zhou
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China
| | - Chengzhi Chen
- Department of Occupational and Environmental Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China.
| | - Jingfu Qiu
- Department of Health Laboratory Technology, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China; Research Center for Environment and Human Health, School of Public Health, Chongqing Medical University, Chongqing 400016, People's Republic of China.
| |
Collapse
|
3
|
Li X, Ma Y, Jiang Q, Zhan H, Sun X. The associations between circulating amino acids and arterial aneurysms and dissection: A bidirectional Mendelian randomization study. INTERNATIONAL JOURNAL OF CARDIOLOGY. CARDIOVASCULAR RISK AND PREVENTION 2025; 25:200388. [PMID: 40160699 PMCID: PMC11951207 DOI: 10.1016/j.ijcrp.2025.200388] [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: 11/28/2024] [Revised: 02/13/2025] [Accepted: 03/05/2025] [Indexed: 04/02/2025]
Abstract
Background Circulating amino acid levels can be altered in arterial aneurysms and dissection, but the relationships between them is unclear. The present study investigated the causal relationship between circulating amino acid levels and arterial aneurysms and dissection via bidirectional two-sample Mendelian randomization (MR). Methods A bidirectional two-sample MR analysis was used. Forward analysis was performed with amino acid levels as the exposure and arterial aneurysms and dissection as outcomes. Reverse analysis was performed with arterial aneurysms and dissection as exposures and circulating amino acid levels as outcomes. MR data were analyzed using five analytical methods: the inverse-variance weighted (IVW), MR‒Egger, weighted median, simple, and weighted methods. IVW was used as the main analytical method, and the other methods were used for supplementary analyses. Heterogeneity was assessed using Cochran's Q test, and horizontal pleiotropy was assessed using intercepts from MR‒Egger regression. The genome-wide association study (GWAS) data for circulating amino acids were obtained from the IEU open GWAS database and the GWAS Catalog database. The GWAS data for arterial aneurysms and dissection were obtained from the Finngen consortium database version R10. Results The tyrosine level was negatively correlated with other aneurysms (P = 0.00211, OR: 0.57, 95 % CI: 0.40, 0.82). Aortic dissection decreased the circulating glycine level (P = 0.00168, OR: 0.98, 95 % CI: 0.98, 0.99). Conclusion Through bidirectional MR analysis, we found that tyrosine level was negatively correlated with other aneurysms and that aortic dissection reduced circulating glycine. Our findings support a possible interaction between circulating amino acid levels and arterial aneurysms and dissection.
Collapse
Affiliation(s)
- Xiaodong Li
- Department of Interventional Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yarong Ma
- Department of Ophthalmology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Qiulin Jiang
- Department of Interventional Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Huizhi Zhan
- Department of Interventional Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Xiaolei Sun
- Department of Interventional Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Department of General Surgery (Vascular Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Laboratory of Nucleic Acids in Medicine for National High-level Talent, Nucleic Acid Medicine of Luzhou Key Laboratory, Southwest Medical University, Luzhou, 646000, China
- Key Laboratory of Medical Electrophysiology, Ministry of Education & Medical Electrophysiological Key Laboratory of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, 646000, China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, 646000, China
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre of Research Excellence, Faculty of Life Science and Medicine, King's College London, London, SE5 9NU, United Kingdom
| |
Collapse
|
4
|
Wang L, Quan W, Song J, Qin Y, Zeng H, Zhang J, Zhao X, Li J, Chen J. Association Between ω-3, ω-6 Polyunsaturated Fatty Acid and Sleep Disorders: From Cross-Sectional to Mendelian Randomization Studies. Food Sci Nutr 2025; 13:e70311. [PMID: 40433111 PMCID: PMC12108440 DOI: 10.1002/fsn3.70311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Sleep disorders are a common health problem affecting a significant proportion of the adult population. Emerging evidence suggests that dietary factors, particularly polyunsaturated fatty acid (PUFA) intake, may play a role in modulating sleep quality. This study aims to investigate the association between omega-3 (ω-3) and omega-6 (ω-6) PUFA and sleep disorders using cross-sectional survey data and data from genome-wide association studies (GWAS). Using data from the National Health and Nutrition Examination Survey (NHANES, 2005-2018), we analyzed a cohort of 31,920 participants, with the primary independent variables being intake of ω-3 and ω-6 PUFAs. Multivariate regression was used to assess associations, and restricted cubic spline analysis was used to explore potential non-linear dose-response relationships. Two-sample Mendelian randomization (MR) analyses were performed to evaluate the causal effects of levels of multiple fatty acids on the risk of sleep disorders. For analysis on NHANES data, the participants with sleep disorders had significantly lower ω-3 PUFA intake (1.71 ± 1.11 g) compared to those without sleep disorders (1.78 ± 1.14 g, p < 0.001). Regression analysis revealed that higher ω-3 PUFA intake was associated with a reduced risk of sleep disorders, while the MR analyses showed that a higher ratio of ω-6 to total fatty acid levels was causally associated with a lower risk of sleep disorders (IVW OR = 0.930, 95% CI: 0.880-0.983, p = 0.011). Our findings suggest that increased ω-3 FA intake and increased ratio of ω-6 to total fatty acid level may be associated with a lower risk of sleep disorders, highlighting the potential benefits of dietary modification for sleep health. Future research should further explore these associations and consider intervention studies to establish causality and optimal dietary recommendations to prevent sleep disorders.
Collapse
Affiliation(s)
- Lin Wang
- Department of NeurologyChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Wei Quan
- Department of NeurologyChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Jia Song
- Department of NeurologyChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Yidan Qin
- Department of NeurologyChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Huibin Zeng
- Department of NeurologyChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Jian Zhang
- Department of NeurologyChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Xuan Zhao
- Department of NeurologyChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Jia Li
- Department of NeurologyChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| | - Jiajun Chen
- Department of NeurologyChina‐Japan Union Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
5
|
Cummings BB, Joing MP, Bouchard PR, Milton MN, Moesta PF, Ramanan V, Lane A, Hirman J, Trauger JW, Maratos-Flier E, Voznesensky A, Splawski I, Nimonkar AV, Flaherty MM, Yi BA, Meyers D, Huet F, Sahambi SK, Yates DP, Hom D, Hinder M, Basson CT, O'Donnell C, Siegelman ES, Garrett CE, Lehrer-Graiwer J, Juliano RA, Weiss EJ. Safety and efficacy of a novel ANGPTL4 inhibitory antibody for lipid lowering: results from phase 1 and phase 1b/2a clinical studies. Lancet 2025; 405:1923-1934. [PMID: 40383129 DOI: 10.1016/s0140-6736(25)00825-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/16/2025] [Accepted: 04/23/2025] [Indexed: 05/20/2025]
Abstract
BACKGROUND Genetic studies have established angiopoietin-related protein 4 (ANGPTL4) as a key regulator of triglyceride metabolism and a promising target to reduce atherosclerotic cardiovascular disease (ASCVD) risk beyond traditional risk factors. Human ANGPTL4 loss-of-function shows no adverse consequences and is associated with reduced triglycerides and remnant cholesterol, and a reduced risk of type 2 diabetes and ASCVD. Nonetheless, development of ANGPTL4 inhibitors has been delayed due to adverse findings in ANGPTL4-knockout mice fed a high saturated fat diet, including lipid accumulation in mesenteric lymph nodes, systemic inflammation, adverse clinical signs, and reduced survival. We previously reported the development and preclinical characterisation of MAR001, an ANGPTL4 inhibitory antibody. Here, we report a comprehensive safety assessment of ANGPTL4 inhibition, including novel analysis of genetic ANGPTL4 loss on mesenteric lymph node architecture in humans and two early-phase clinical trials. METHODS MAR001 was evaluated in a first-in-human, randomised, placebo-controlled, single-ascending-dose phase 1 study with three parts in which participants received a single subcutaneous injection of MAR001 or placebo. The study was developed and conducted by Novartis Biomedical Research (Cambridge, MA, USA). Eligible participants enrolled in part 1A were healthy men and women aged between 18 years and 65 years with a bodyweight of at least 50 kg and a BMI of 18-30 kg/m2. Participants in part 1B weighed at least 70 kg and had a BMI of 30-40 kg/m2. Participants in part 1C weighed at least 59 kg and had fasting triglycerides in the range of 200-500 mg/dL. The primary objectives were to assess the safety and tolerability of a single subcutaneous injection of MAR001 up to and including 141 days post-dose and to assess the pharmacokinetics of single-dose subcutaneous administration in healthy participants. MAR001 was subsequently assessed in a randomised, double-blind, placebo-controlled phase 1b/2a study in participants with metabolic dysfunction. The study was done at two sites in Australia. Eligible participants were adults with hypertriglyceridaemia (in the screening range of ≥1·7 mmol/L and ≤5·6 mmol/L; ≥151 mg/dL and ≤496 mg/dL) and a history of type 2 diabetes, or a screening homeostatic model assessment for insulin resistance (HOMA-IR) value greater than 2·2 and abdominal obesity (defined as waist circumference >88 cm for women and >102 cm for men; > 80 cm for Asian women and >90 cm for Asian men). The primary objective was to characterise the safety and tolerability of multiple doses of MAR001 in participants with metabolic dysfunction. The phase 1b/2a study is registered with ClinicalTrials.gov, NCT05896254. FINDINGS We found no evidence of clinical adversity in human germline ANGPTL4 loss-of-function, adding to preclinical support for initiating human studies. Between Nov 20, 2017, and Sept 10, 2019, in the first-in-human, randomised, placebo-controlled, single-ascending-dose phase 1 study, part 1A enrolled 32 healthy participants: six each received 15 mg, 50 mg, 150 mg, or 450 mg of MAR001, and eight received placebo. Part 1B enrolled 12 participants: nine received 450 mg of MAR001 and three received placebo. Part 1C enrolled 12 participants: eight received 450 mg of MAR001 and four received placebo. Between Nov 24, 2013, and July 1, 2024, in the multidose phase 1b/2a randomised, double-blind, placebo-controlled study, 55 participants were randomly assigned to receive subcutaneous injections of placebo (19 participants) or MAR001 at doses of 150 mg (ten participants), 300 mg (nine participants), or 450 mg (17 participants), followed by a 12-week safety follow-up period. MAR001 was safe and generally well tolerated, and we observed no treatment-related systemic inflammatory biomarker elevations or changes in mesenteric lymph node size or inflammation assessed by MRI. MAR001 (450 mg) yielded placebo-adjusted week 12 mean reductions in triglycerides of 52·7% (90% CI -77·0 to -28·3) and in remnant cholesterol of 52·5% (-76·1 to -28·9). INTERPRETATION ANGPTL4 inhibition with MAR001 can safely and effectively reduce circulating triglycerides and remnant cholesterol. The findings of these trials support further research and development of MAR001 as a promising potential lipid-lowering therapy to reduce risk of ASCVD. FUNDING Marea Therapeutics.
Collapse
Affiliation(s)
| | | | - Page R Bouchard
- Marea Therapeutics, San Francisco, CA, USA; Novartis Biomedical Research, Cambridge, MA, USA
| | - Mark N Milton
- Marea Therapeutics, San Francisco, CA, USA; Novartis Biomedical Research, Cambridge, MA, USA
| | | | | | | | - Joe Hirman
- Marea Therapeutics, San Francisco, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | - Douglas Hom
- Novartis Biomedical Research, Cambridge, MA, USA
| | | | | | | | - Evan S Siegelman
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | | |
Collapse
|
6
|
Fan H, Song H, Fu Q, Du G, Bai Y, Zhang C, Shi X. Bidirectional mendelian randomization assessment of causality between lactate levels and multiple autoimmune diseases. Sci Rep 2025; 15:17700. [PMID: 40399513 PMCID: PMC12095665 DOI: 10.1038/s41598-025-02507-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 05/13/2025] [Indexed: 05/23/2025] Open
Abstract
Bidirectional two-sample Mendelian randomization (MR) was performed to provide genetic evidence for the causal association between multiple sclerosis (MS), type 1 diabetes(T1D), rheumatoid arthritis (RA) and inflammatory bowel disease (IBD), Crohn's disease (CD), systemic lupus erythematosus (SLE), ulcerative colitis (UC) and lactate levels. Inverse variance weighted (IVW), weighted median estimator (WME), weighted mode, and MR-Egger regression were used to assess the potential causal links. Sensitivity analysis included Cochran's Q test for heterogeneity, Steiger test of directionality for directionality, MR-Egger regression, MR pleiotropy residual sum and outlier (MR-PRESSO), and leave-one-out method. MR analysis utilized 510 SNPs associated with seven different kinds of autoimmune diseases and 11 SNPs associated with lactate levels as IVs. No significant genetic association between any autoimmune diseases and lactate levels was discovered by IVW. While IVW revealed no significant associations, exploratory analyses using WME and weighted mode methods identified nominal links between RA/IBD and lactate levels (RA: WME OR = 1.01, P = 0.010; weighted mode OR = 1.01, P = 0.008; IBD: weighted mode OR = 1.01, P = 0.042). These findings, though not surviving FDR correction, warrant further investigation. In reverse MR analysis, there was no significant association between lactate level exposure and any autoimmune disease outcomes. MR-Egger regression indicated potential horizontal pleiotropy in the RA-lactate analysis and Cochran's Q test suggest no absence of heterogeneity. Potential reverse causality in the analysis of SLE as outcomes and lactate levels as exposures was discovered by MR Steiger. Based on limited evidence, Our MR Analysis found a possible genetic causal association between RA and lactate level difference.
Collapse
Affiliation(s)
- Hua Fan
- Office of Research & Innovation, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, No. 25, Jinghua Road, Jianxi District, Luoyang, 471003, Henan, China.
- Luoyang Key Laboratory of Neuroimmunology and innovative Drug Screening, Luoyang, 471003, Henan, China.
| | - Hongna Song
- Department of Neuro-auricular vertigo, The Second Affiliated Hospital of Henan, University of Science and Technology, Luoyang, 471003, Henan, China
| | - Qizhi Fu
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan, University of Science and Technology, Luoyang, 471003, Henan, China
| | - Ganqin Du
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan, University of Science and Technology, Luoyang, 471003, Henan, China
| | - Yongjie Bai
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan, University of Science and Technology, Luoyang, 471003, Henan, China
| | - Canfei Zhang
- Department of Neurology, The First Affiliated Hospital, College of Clinical Medicine of Henan, University of Science and Technology, Luoyang, 471003, Henan, China
| | - Xiaofei Shi
- Department of Rheumatology and Immunology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, No. 25, Jinghua Road, Jianxi District, Luoyang, 471003, Henan, China.
| |
Collapse
|
7
|
Lu J, Li GHY, Hu J, Wang Z. Genetic insights support PARP1 as a mediator in the protective association of ATP-citrate lyase inhibitors with melanoma. Commun Biol 2025; 8:777. [PMID: 40399559 PMCID: PMC12095509 DOI: 10.1038/s42003-025-07860-z] [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: 08/25/2024] [Accepted: 02/28/2025] [Indexed: 05/23/2025] Open
Abstract
ATP-citrate lyase (ACLY) inhibitors emerge as a promising anti-cancer strategy, yet their causal effects across various cancer types remain unclear. Here, we employ a drug-target Mendelian randomization (MR) approach using four cis-expression quantitative trait loci for blood ACLY gene expression as genetic instruments to mimic ACLY inhibition. We utilize genetic data from the eQTLGen consortium (N = 31,684) for ACLY expression, the deCODE study (N = 35,559) for plasma proteome, and large-scale cancer genome-wide association studies consortia (N from 49,708 to 417,127) to investigate the association of genetically mimicked ACLY inhibitors with 17 cancers and identify potential mediating proteins. Genetically proxied ACLY inhibition is strongly associated with reduced melanoma risk (odds ratio [95% confidence interval (CI)]: 0.85 [0.78, 0.92]) in a combined analysis of two independent outcome datasets. Proteome-wide MR screening 1517 plasma proteins identifies 3 proteins associated with melanoma, with Poly [ADP-ribose] polymerase 1 (PARP1) showing strong colocalization support. Mediation analysis further suggests PARP1 as a mediator in the protective effect of ACLY inhibition on melanoma (mediated proportion [95% CI]: 51.52% [5.45%, 97.58%]). Follow-up and validation analyses support the robustness of these results. This study illuminates the therapeutic potential of ACLY inhibition in melanoma, with PARP1 implicated as a potential mediator, offering avenues for targeted interventions.
Collapse
Affiliation(s)
- Jiawen Lu
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hong Kong, China
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China
| | - Gloria Hoi-Yee Li
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hong Kong, China
| | - Jingyi Hu
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology, Ministry of Education, Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, China
| | - Zhenqian Wang
- Department of Health Technology and Informatics, Faculty of Health and Social Sciences, The Hong Kong Polytechnic University, Hong Kong, China.
- School of Public Health (Shenzhen), Sun Yat-sen University, Shenzhen, China.
| |
Collapse
|
8
|
Kim BR, Kim G, Jin SP, Choi CW, Kim J, Park H. Causal association between polyunsaturated fatty acids and acne: a two-sample Mendelian randomization study. Br J Dermatol 2025; 192:1106-1114. [PMID: 39936505 DOI: 10.1093/bjd/ljaf052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/03/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND Observational studies have demonstrated a close association between polyunsaturated fatty acids (PUFAs) and acne. However, the findings of clinical trials have been inconsistent, leaving the causal relationship between PUFAs and acne unclear. OBJECTIVES To investigate the causal association between genetically proxied PUFAs and acne risk. METHODS Mendelian randomization (MR) was performed using single nucleotide polymorphisms associated with PUFAs as instrumental variables. The causal associations between PUFAs and acne were estimated among 115 006 UK Biobank participants and 363 927 participants of Finnish descent. RESULTS Genetically predicted docosahexaenoic acid (DHA) levels [β = -0.303, 95% confidence interval (CI) -0.480 to -0.126; P = 7.74 × 10-4] and its percentage to total fatty acids (β = -0.402, 95% CI -0.651 to -0.258; P = 5.91 × 10-6) showed a significant causal association with a decreased risk of acne. Conversely, genetically predicted percentages of linoleic acid (LA) in total fatty acids (β = 0.768, 95% CI 0.411-0.126; P = 2.87 × 10-4) and omega-6 : omega-3 ratio (β = 0.373, 95% CI 0.142-0.604; P = 4.48 × 10-3) were robustly associated with an increased risk of acne. These effects were attenuated after excluding a genetic variant of rs174528 located upstream of FADS1, highlighting the biologic link between FADS1 and delta-5 desaturase activity. Multivariable MR analysis indicated that PUFAs were causally associated with acne, independent of body mass index. CONCLUSIONS Our study indicates that high DHA levels and their ratios to total fatty acids have causal protective effects against acne, while high LA levels and omega-6 : omega-3 ratio are associated with increased acne risk. This association was largely attributable to the influence of genetic variants related to FADS1.
Collapse
Affiliation(s)
- Bo Ri Kim
- Department of Dermatology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Gahyun Kim
- Precision Medicine Center, Future Innovation Research Division, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Seon-Pil Jin
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Dermatology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Chong Won Choi
- Department of Dermatology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jinho Kim
- Precision Medicine Center, Future Innovation Research Division, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Genomic Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Hyunsun Park
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Dermatology, Seoul National University Hospital, Seoul, Republic of Korea
| |
Collapse
|
9
|
Li XC, Wang B, Tang Y. Causal relationships between four types of lipids and breast cancer risk with potential mediators: evidence from Mendelian randomization study and bioinformatics analysis. Discov Oncol 2025; 16:791. [PMID: 40379931 PMCID: PMC12084463 DOI: 10.1007/s12672-025-02597-x] [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: 02/04/2025] [Accepted: 05/06/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND AND PURPOSE Breast cancer (BC) is the primary cause of cancer-related deaths among women worldwide, with increasing evidence pointing to the effect of metabolic factors, particularly lipid levels, in its pathogenesis. In this research, Mendelian randomization (MR) was employed to explore the causality between four plasma lipid traits-total cholesterol (TC), triglycerides (TG), high-density lipoprotein cholesterol (HDL-C) and low-density lipoprotein cholesterol (LDL-C)-and the risk of BC. Additionally, we explored the potential mediating effects of coronary artery disease (CAD), total testosterone (TT) on these associations and possible mechanisms through bioinformatics analyses. METHODS Data of genome-wide association study (GWAS) on lipids, CAD, TT and BC were obtained from public sources and websites as part of a genome-wide association research. The inference of causality was primarily assessed through the inverse variance weighting (IVW) approach, with supplementary tests for horizontal pleiotropy and heterogeneity. To verify the directionality of causal relationships, the MR Steiger test was applied. Additionally, reverse causality was evaluated by regarding BC as the exposure. To adjust for confounders, multivariate MR (MVMR) was performed, followed by a two-step mediation analysis to investigate the mediating roles of CAD in the lipid-BC association, and of TT in the CAD-BC relationship. The intersecting SNP (rs11556924) between causal pathways was established through a Venn diagram and its associated gene (Zinc Finger C3HC-Type Containing 1, ZC3HC1) was identified through the g:Profiler database. The expression of ZC3HC1 was further explored using the TIMER, GEPIA2 and HPA database. Finally, enrichment analyses of Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) and protein-protein interactions (PPI) network analysis were conducted on ZC3HC1 and its related genes. RESULTS The random-effects IVW analysis yielded the following results: HDL-C on CAD (OR = 0.843, 95% CI 0.771-0.921, P < 0.001), CAD on BC (OR = 0.935, 95% CI 0.892-0.980, P = 0.005), HDL-C on BC (OR = 1.127, 95% CI 1.059-1.199, P < 0.001), CAD on TT (OR = 0.987, 95% CI 0.975-0.998, P = 0.020) and TT on BC (OR = 1.354, 95% CI 1.148-1.598, P < 0.001). The MR Steiger test results support the validity of the inferred causal direction (P < 0.001). There were no discernible causal relationships between BC and HDL-C/CAD according to reverse MR analysis (P > 0.05). Following MVMR adjustment, the causal effects of HDL-C, CAD, and TT on BC were still statistically significant (P < 0.05). Besides, the two-step mediation analysis indicated that CAD mediated 7.8% of the causal effect of HDL-C on BC, whereas TT mediated 6.1% of the causal effect between CAD and BC. The expression of ZC3HC1 showed no significant expression difference between normal and BC tissues (P > 0.05), which might indicate a carcinogenic effect independent of expression levels but driven by functional alterations induced by variants (C > T). Functional network analysis suggested that ZC3HC1 was associated with multiple signal pathways in cancers, such as PI3K-Akt and MAPK signal pathways. CONCLUSIONS From a genetic perspective, our study reveals that there is causality between HDL-C levels and BC risk, with CAD and TT acting as partial mediators in this relationship. Moreover, our study firstly establishes a potential link between CAD-associated SNP (rs11556924), the corresponding gene (ZC3HC1) functional dysregulation, and the initiation of BC. These findings shed light on the biological links between lipids and BC, potentially contributing to future prevention and treatment strategies.
Collapse
Affiliation(s)
- Xu-Chu Li
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 South Panjiayuan Lane, Chaoyang District, Beijing, 100021, China
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Bangqi Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yu Tang
- Clinical Trial Center, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, 17 South Panjiayuan Lane, Chaoyang District, Beijing, 100021, China.
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
10
|
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
|
11
|
Cummings BB, Bouchard PR, Milton MN, Moesta PF, Ramanan V, Trauger JW, Maratos-Flier E, Voznesensky A, Splawski I, Nimonkar AV, DiPetrillo K, LaSala D, Pan M, Flaherty MM, Huet F, Sahambi SK, Dong J, Knee D, Cebe R, Huber T, Lehrer-Graiwer J, Juliano RA, Weiss EJ. An ANGPTL4 inhibitory antibody safely improves lipid profiles in non-human primates. EBioMedicine 2025:105748. [PMID: 40360349 DOI: 10.1016/j.ebiom.2025.105748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Revised: 04/23/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Angiopoietin-like protein 4 (ANGPTL4) inhibition is a promising approach to manage atherogenic dyslipidaemia and residual atherosclerotic cardiovascular disease (ASCVD) risk. Human ANGPTL4 loss-of-function (LoF) is associated with reduced plasma triglyceride (TG), remnant cholesterol (RC), and apolipoprotein B (ApoB) levels, and lower risk of type 2 diabetes and ASCVD, without observable safety concerns. However, development of ANGPTL4 inhibitors has been stalled by adverse findings in Angptl4 knockout mice fed a high-saturated-fat diet (HSFD), which show lipid accumulation in mesenteric lymph nodes (MLNs), systemic inflammation, severe adverse clinical signs, and reduced survival. METHODS Here, we present the development and preclinical characterisation of MAR001, a humanised monoclonal ANGPTL4 inhibitor antibody. We assessed single-dose MAR001 efficacy in hypertriglyceridemic (HTG) non-human primates (NHPs, n = 4), and safety in two NHP toxicology studies: a 15-week subchronic study with a standard or HSFD (n = 36), and a 9-month chronic study exclusively on an HSFD (n = 24). FINDINGS In HTG monkeys, single-dose MAR001 treatment reduced plasma TG by up to 58%, non-high-density lipoprotein cholesterol by 38%, ApoB by 30%, and RC by 59%. In safety studies, MAR001 was well tolerated without clinically adverse findings with either diet. Animals fed an HSFD exhibited minimal to moderate foamy macrophage formation in MLNs, but importantly, these histological findings did not progress to degeneration, necrosis, inflammation, fibrosis, or other reactive changes, and with no evidence of systemic effects, including no evidence of systemic inflammation or clinical adverse signs. INTERPRETATION MAR001 improved plasma lipid profiles in NHPs without clinical adversity, even during prolonged HSFD feeding. The favourable NHP safety profile aligns with human ANGPTL4 LoF findings, and contrasts with the severe pathology in mouse knockout models on an HSFD. These findings supported MAR001 clinical studies reported in our concurrent publication, which demonstrated robust lipid improvements without lymphatic pathology. Overall, these findings support continued development of MAR001 as a promising new therapy for ASCVD risk reduction. FUNDING Marea Therapeutics.
Collapse
Affiliation(s)
- Beryl B Cummings
- Marea Therapeutics, 131 Oyster Point Boulevard, South San Francisco, CA, 94080, USA
| | - Page R Bouchard
- Marea Therapeutics, 131 Oyster Point Boulevard, South San Francisco, CA, 94080, USA; Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Mark N Milton
- Marea Therapeutics, 131 Oyster Point Boulevard, South San Francisco, CA, 94080, USA; Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Peter F Moesta
- Marea Therapeutics, 131 Oyster Point Boulevard, South San Francisco, CA, 94080, USA
| | - Vyas Ramanan
- Marea Therapeutics, 131 Oyster Point Boulevard, South San Francisco, CA, 94080, USA
| | - John W Trauger
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | | | - Andrei Voznesensky
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Igor Splawski
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Amitabh V Nimonkar
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Keith DiPetrillo
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Daniel LaSala
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Meihui Pan
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Meghan M Flaherty
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Francois Huet
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Sukhdeep K Sahambi
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Jijun Dong
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Deborah Knee
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Regis Cebe
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | - Thomas Huber
- Novartis Institutes for Biomedical Research, 22 Windsor Street, Cambridge, MA 02139, USA
| | | | - Rebecca A Juliano
- Marea Therapeutics, 131 Oyster Point Boulevard, South San Francisco, CA, 94080, USA
| | - Ethan J Weiss
- Marea Therapeutics, 131 Oyster Point Boulevard, South San Francisco, CA, 94080, 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
|
Gao X, Zhang W, Wang F. Circulating branched-chain amino acids and risk of psychiatric disorders: A two-sample Mendelian randomization study. J Psychosom Res 2025; 192:112101. [PMID: 40107167 DOI: 10.1016/j.jpsychores.2025.112101] [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: 08/14/2024] [Revised: 03/08/2025] [Accepted: 03/12/2025] [Indexed: 03/22/2025]
Abstract
OBJECTIVE Several studies have identified biomarkers that help in the prevention, diagnosis, and treatment of psychiatric disorders. The current study aimed to explore the association between circulating branched-chain amino acids (BCAA) and the risk of five psychiatric disorders. METHODS GWAS summary statistics were obtained from the UK Biobank and the FinnGen Biobank. The inverse variance weighted (IVW) method was used as a primary method to assess causal effects. The heterogeneity test, the horizontal pleiotropy test, and leave-one-out analysis were used to assess the robustness and reliability of the results. RESULTS Total BCAA levels were not significantly associated with the risk of anxiety disorders, alcohol dependence, depression, panic disorder, and bipolar affective disorders. Separate analyses for each BCAA showed that the levels of valine and leucine were not associated with the risk of the mentioned five psychiatric disorders, and the levels of isoleucine were not associated with the risk of depression, panic disorder, and bipolar affective disorders, but with the risk of anxiety disorders (IVW: OR = 0.814, 95 %CI = 0.716-0.925) and alcohol dependence (IVW: 0.690, 95 %CI = 0.560-0.849). CONCLUSIONS Circulating isoleucine was a protective factor for anxiety disorders and alcohol dependence, offering a new research direction and theoretical basis for preventing and managing psychiatric disorders.
Collapse
Affiliation(s)
- Xueren Gao
- School of Pharmacy, Yancheng Teachers' University, Yancheng, Jiangsu, China.
| | - Weichao Zhang
- School of Pharmacy, Yancheng Teachers' University, Yancheng, Jiangsu, China
| | - Fang Wang
- School of Pharmacy, Yancheng Teachers' University, Yancheng, Jiangsu, China
| |
Collapse
|
14
|
Xue CC, Li H, Yu M, Chong CCY, Fan Q, Tham YC, Cheung CMG, Wong TY, Chew EY, Cheng CY. Omega-3 Fatty Acids as Protective Factors for Age-Related Macular Degeneration: Prospective Cohort and Mendelian Randomization Analyses. Ophthalmology 2025; 132:598-609. [PMID: 39662686 DOI: 10.1016/j.ophtha.2024.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/28/2024] [Accepted: 12/03/2024] [Indexed: 12/13/2024] Open
Abstract
PURPOSE Epidemiologic studies and clinical trials have reported inconsistent findings regarding omega-3 fatty acids' protective role in age-related macular degeneration (AMD). We investigated their association in a prospective cohort and examined causality using Mendelian randomization (MR) analyses. DESIGN Prospective cohort study and 2-sample MR analyses. PARTICIPANTS The cohort included 258 350 AMD-free individuals of European descent from the UK Biobank. Mendelian randomization analyses used genome-wide association study data on plasma omega-3 and docosahexaenoic acid (DHA) (UK Biobank, n = 115 006) and AMD (dry, wet, and any; FinnGen, n = 208 690-209 122). METHODS Cox regression assessed the association between plasma omega-3 and DHA levels and AMD incidence, adjusting for systemic covariates and AMD polygenetic risk score (PRS). Interaction effects of AMD genetic risk (PRS, complement factor H and age-related maculopathy susceptibility 2 genotypes), and plasma omega-3 and DHA levels were tested. For MR analyses, we used random-effect inverse-variance weighted model as primary, with 5 sensitivity models. Causality was considered significant if P < 0.05 in the primary model and at least 2 sensitivity models. MAIN OUTCOME MEASURES Risk of AMD. RESULTS Over 12.9 years, 5068 people (1.9%) demonstrated AMD. Higher plasma levels (in millimoles per liter) of omega-3 (hazard ratio [HR], 0.80; 95% confidence interval [CI], 0.72-0.95; P = 0.006) and DHA (HR, 0.65; 95% CI, 0.44-0.96; P = 0.029) were associated with lower risk of receiving an AMD diagnosis. Mendelian randomization showed genetic predisposition to higher plasma omega-3 levels reduced the risk of dry AMD (odds ratio [OR], 0.83; 95% CI, 0.73-0.96; P = 0.010), wet AMD (OR, 0.76; 95% CI, 0.65-0.88; P < 0.001), and any AMD (OR, 0.82; 95% CI, 0.74-0.92; P < 0.001). Similar results were found for plasma DHA levels (wet AMD:OR, 0.79; 95% CI, 0.65-0.96; P = 0.017; any AMD: OR, 0.84; 95% CI, 0.72-0.98; P = 0.030). No significant interaction was found between omega-3 and DHA levels and AMD genetic risk (all P > 0.05). CONCLUSIONS Both the prospective and MR analyses suggest omega-3 and DHA may protect against AMD, supporting the need for further clinical trials to test their effectiveness in AMD prevention and treatment. FINANCIAL DISCLOSURE(S) Proprietary or commercial disclosure may be found in the Footnotes and Disclosures at the end of this article.
Collapse
Affiliation(s)
- Can Can Xue
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Republic of Singapore
| | - Hengtong Li
- Centre for Innovation and Precision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore; Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore
| | - Marco Yu
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Republic of Singapore
| | - Crystal Chun Yuen Chong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Republic of Singapore
| | - Qiao Fan
- Centre for Quantitative Medicine, Duke-NUS Medical School, Singapore, Republic of Singapore
| | - Yih-Chung Tham
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Republic of Singapore; Centre for Innovation and Precision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore; Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore; Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Republic of Singapore
| | - Chui Ming Gemmy Cheung
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Republic of Singapore; Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Republic of Singapore
| | - Tien Yin Wong
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Republic of Singapore; School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua Medicine, Tsinghua University, Beijing, China
| | - Emily Y Chew
- Division of Epidemiology and Clinical Applications, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Ching-Yu Cheng
- Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, Republic of Singapore; Centre for Innovation and Precision Eye Health, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore; Department of Ophthalmology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Republic of Singapore; Ophthalmology & Visual Sciences Academic Clinical Program (Eye ACP), Duke-NUS Medical School, Singapore, Republic of Singapore.
| |
Collapse
|
15
|
Yuan F, Jia G, Wen W, Xu S, Gunchick V, Deng K, Long J, Yu D, Shu XO, Zheng W. Blood metabolic biomarkers and colorectal cancer risk: results from large prospective cohort and Mendelian randomisation analyses. Br J Cancer 2025:10.1038/s41416-025-02997-4. [PMID: 40307439 DOI: 10.1038/s41416-025-02997-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 03/04/2025] [Accepted: 03/21/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Emerging evidence suggests metabolic dysregulation may contribute to colorectal cancer (CRC) aetiology. We aimed to identify pre-diagnostic metabolic biomarkers for CRC risk in 230,420 UK Biobank participants. METHODS Nuclear magnetic resonance spectroscopy was used to quantify 249 metabolic biomarkers in plasma samples collected at baseline. Cox proportional hazards models were used to estimate hazard ratios and 95% confidence intervals (CIs) for associations of metabolic biomarkers with CRC risk after adjusting for potential confounders. To infer the potential causality of biomarkers that were associated with CRC independent of the others, we performed genome-wide association analyses among 199,732 UK Biobank participants of European ancestry to identify biomarker-associated genetic variants, followed by two-sample Mendelian randomization (MR) analyses using summary statistics of 78,473 CRC cases and 107,143 controls of European ancestry. RESULTS During a median follow-up time of 9.7 years, 2,410 incident primary CRC cases were identified. Among 43 CRC-associated (P-value < 0.001) metabolic biomarkers, ten biomarkers including fatty acids (FAs), inflammation, ketone bodies, and lipoprotein lipids were associated with CRC risk after mutual adjustment. MR analyses provided strong evidence for potential causal associations of CRC risk with percentages of linolic acid [odds ratio (OR) = 0.89, 95% CI = 0.83-0.96, P-value = 3 × 10-3] and saturated FAs (OR = 1.14, 95% CI = 1.03-1.25, P-value = 9 × 10-3) to total FAs. CONCLUSIONS We identified multiple CRC-associated metabolic biomarkers. Perturbed lipid and lipoprotein metabolism may promote colorectal carcinogenesis.
Collapse
Affiliation(s)
- Fangcheng Yuan
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Guochong Jia
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wanqing Wen
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shuai Xu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Valerie Gunchick
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kui Deng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jirong Long
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Danxia Yu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wei Zheng
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
16
|
Wang L, Ding Y, Tang Y, Yang M, Yang Z, Yang X, Xia J. Association between sphingomyelin levels and gut microbiota abundance in Alzheimer's disease: a two-sample Mendelian randomization study. BMC Neurol 2025; 25:191. [PMID: 40307740 PMCID: PMC12044981 DOI: 10.1186/s12883-025-04207-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 04/24/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Several previous observational studies have shown that abnormal sphingomyelin metabolism may be implicated in the pathogenesis of Alzheimer's disease. To determine the causal relationship between sphingolipid abundance and gut microbiota abundance at the genetic level, we conducted a Mendelian randomization (MR) investigation. METHODS We first used the TwoSampleMR and MRPRESSO packages for conducting two-sample MR studies. Second, we utilized random effect inverse variance weighting (IVW) as the principal method of analysis and used MR‒Egger, the weighted median, the simple mode and the weighted mode as supplementary methods. Finally, we performed tests for heterogeneity and horizontal pleiotropy. These analyses were also conducted to evaluate the impact of individual SNPs on the outcomes of our analysis. A Bonferroni-corrected threshold of p = 2.4e-4(0.05/211) was considered significant, and p values less than 0·05 were considered to be suggestive of an association. RESULTS The results showed that sphingolipid levels were suggestively associated with the abundance of 6 gut microbiota taxa. Specifically, two taxa were positively correlated with sphingolipid levels, including the family Alcaligenaceae (p = 0.006, OR 95% CI = 1.109 [1.030-1.194]) and the species Ruminococcus callidus (p = 0.034, OR 95% CI = 1.217 [1.015-1.460]). In contrast, negative correlations were observed with the abundances of 4 gut microbiota taxa, including the genus Flavonifractor (p = 0.026, OR 95% CI = 0.804 [0.663-0.974]), the genus Streptococcus (p = 0.014, OR 95% CI = 0.909 [0.842-0.981]), the species Bacteroides caccae (p = 0.037, OR 95% CI = 0.870 [0.763-0.992]), and the species Haemophilus parainfluenzae (p = 0.006, beta 95% CI = -0.269 [-0.462, -0.076]). The results presented a normal distribution, with no anomalous values, heterogeneity, or horizontal pleiotropic effects detected. CONCLUSIONS This two-sample MR study revealed a potential causal relationship between sphingomyelin levels and gut microbiota abundance.
Collapse
Affiliation(s)
- Liping Wang
- Department of General Surgery, Institute of General Surgical Research, Jiangnan University Medical Center, Wuxi, China
| | - Yuyan Ding
- Department of General Surgery, Institute of General Surgical Research, Jiangnan University Medical Center, Wuxi, China
| | - Yu Tang
- Department of General Surgery, Institute of General Surgical Research, Jiangnan University Medical Center, Wuxi, China
| | - Mengqi Yang
- Department of General Surgery, The Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, Wuxi, China
| | - Zhihui Yang
- Department of General Surgery, Institute of General Surgical Research, Jiangnan University Medical Center, Wuxi, China
| | - Xiao Yang
- Department of General Surgery, Institute of General Surgical Research, Jiangnan University Medical Center, Wuxi, China
| | - Jiazeng Xia
- Department of General Surgery, Institute of General Surgical Research, Jiangnan University Medical Center, Wuxi, China.
- Department of General Surgery, The Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, Wuxi, China.
- Wuxi Clinical College, Nantong University, Wuxi, China.
| |
Collapse
|
17
|
Qiu J, Hu P, Li F, Huang Y, Yang Y, Sun F, Wu P, Lai Y, Wang Y, He X, Dong Y, Zhang P, Zhang S, Wu N, Wang T, Yang S, Li S, Yuan J, Liu X, Liu G, Hu Y, Wu JHY, Chen D, Pan A, Pan XF. Circulating linoleic acid and its interplay with gut microbiota during pregnancy for gestational diabetes mellitus. BMC Med 2025; 23:245. [PMID: 40289092 PMCID: PMC12036143 DOI: 10.1186/s12916-025-04061-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Circulating linoleic acid (LA) levels have been reported to be associated with various metabolic outcomes. However, the role of LA and its interplay with gut microbiota in gestational diabetes mellitus (GDM) remains unclear. This study aimed to investigate the longitudinal association between circulating LA levels during pregnancy and the risk of GDM, and the potential role of gut microbiota. METHODS A nested case-control study was conducted within the ongoing Tongji-Huaxi-Shuangliu Birth Cohort in Chengdu, China. Blood and fecal samples were collected during early and middle pregnancy from 807 participants. GDM was diagnosed in middle pregnancy using the International Association of Diabetes and Pregnancy Study Groups criteria. Plasma LA levels were measured using gas chromatography-mass spectrometry, and gut microbiota was analyzed through 16S rRNA gene sequencing and shotgun metagenomic sequencing. A two-sample Mendelian randomization study was conducted using data from the IEU OpenGWAS database and the FinnGen consortium. RESULTS Elevated plasma LA levels were associated with a lower risk of GDM in both early (P for trend = 0.002) and middle pregnancy (P for trend = 0.02). Consistently, Mendelian randomization analysis revealed that each unit increase in LA was associated with a 16% reduction in GDM risk (odds ratio: 0.84, 95% confidence interval: 0.72, 0.95). In early pregnancy, higher plasma LA levels were correlated with higher adiponectin levels (P < 0.001) and lower levels of triglycerides (P < 0.001), HbA1c (P = 0.04), and C-peptide (P = 0.04). The LA-accociated microbiota mediated the relationship between LA and C-peptide (P = 0.01). Additionally, the inverse association between LA and GDM was modified by Bilophila (P for interaction = 0.03), with a stronger association observed in participants with lower Bilophila levels in early pregnancy. Metagenomic analyses further showed that the LA-associated pathway (D-galacturonate degradation I) and its key enzyme (EC 4.2.1.7) were associated with metabolic traits. CONCLUSIONS Our study provides evidence for an inverse causal association between plasma LA levels during pregnancy and GDM risk, which is both mediated and modified by gut microbiota.
Collapse
Affiliation(s)
- Jiahui Qiu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology & Section of Epidemiology and Population Health, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ping Hu
- Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Fan Li
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yichao Huang
- Department of Toxicology, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Yunhaonan Yang
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fengjiang Sun
- School of Environment and Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou, Guangdong, China
| | - Ping Wu
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuwei Lai
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yi Wang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xiangwang He
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yidan Dong
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Peiqi Zhang
- Department of Epidemiology and Biostatistics, School of Public Health, Chengdu Medical College, Chengdu, Sichuan, China
| | - Shanshan Zhang
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Nianwei Wu
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Tianlei Wang
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shizhuo Yang
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Shuo Li
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiaying Yuan
- Department of Science and Education & Shuangliu Institute of Women's and Children's Health, Shuangliu Maternal and Child Health Hospital, Chengdu, Sichuan, China
| | - Xiaojuan Liu
- Department of Laboratory Medicine, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Gang Liu
- Department of Nutrition and Food Hygiene, Hubei Key Laboratory of Food Nutrition and Safety, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yayi Hu
- Department of Obstetrics and Gynecology, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jason H Y Wu
- School of Population Health, The George Institute for Global Health, University of New South Wales, Sydney, NSW, Australia
| | - Da Chen
- School of Environment and Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou, Guangdong, China.
| | - An Pan
- Department of Epidemiology and Biostatistics, Ministry of Education Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| | - Xiong-Fei Pan
- Section of Epidemiology and Population Health & Department of Gynecology and Obstetrics, Ministry of Education Key Laboratory of Birth Defects and Related Diseases of Women and Children & Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China.
- Shuangliu Institute of Women's and Children's Health, Shuangliu Maternal and Child Health Hospital, Chengdu, Sichuan, China.
- Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
18
|
Wulff AB, Nordestgaard BG. Genetics of remnant cholesterol. Curr Opin Lipidol 2025:00041433-990000000-00119. [PMID: 40277396 DOI: 10.1097/mol.0000000000000991] [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] [Indexed: 04/26/2025]
Abstract
PURPOSE OF REVIEW Remnant cholesterol is receiving increasing attention as a target to reduce residual atherosclerotic cardiovascular disease (ASCVD) risk in individuals already treated with statins. New therapeutic options as antisense oligonucleotides, small interfering RNA, and monoclonal antibodies allow specific targeting of genes and proteins to counter pathological pathways promoted by these genes. Identifying genetic determinants of remnant cholesterol and relating these to risk of ASCVD is thus an appealing path to identifying and evaluating new and existing drug targets. RECENT FINDINGS Human genetic epidemiology has identified several genetic variants in genes involved in lipoprotein metabolism with effect on plasma concentrations of remnant cholesterol. Lipoprotein lipase (LPL) is central to the metabolism of remnant lipoproteins and plasma concentrations of remnant cholesterol, and several genes, including APOC3, ANGPTL3 and ANGPTL4, whose gene products regulate activity of LPL, are important determinants of remnant cholesterol. SUMMARY Current opinion is that remnant cholesterol is a likely causal factor in the development of ASCVD. Human genetic studies have identified several genes, many involved in LPL function, affecting remnant cholesterol concentrations, some of which are already used as therapeutic targets, and others which are subject to investigation of their remnant cholesterol and triglyceride-lowering effect in clinical trials.
Collapse
Affiliation(s)
- Anders B Wulff
- Department of Clinical Biochemistry
- The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry
- The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
19
|
Zhao S, Liu X, Wang Q, Xiao S, Wang W, Dong X, Jiang Y, Wu W. Dietary factors and oral cancer risk: a comprehensive Mendelian randomization analysis in a European population. Discov Oncol 2025; 16:540. [PMID: 40240665 PMCID: PMC12003238 DOI: 10.1007/s12672-025-02247-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Oral cancer is one of the most prevalent malignant tumors in the field of oral and maxillofacial surgery. The main etiological factors are tobacco and alcohol, and diet is now considered an important factor in its development. However, it remains to be elucidated whether the associations between specific diets and oral cancer risk are causal. Therefore, in this study, we performed two-sample Mendelian randomization (MR) analyses to estimate the associations of specific diets with the risk of oral cavity cancer (OCC). METHODS We conducted a two-sample MR analysis to assess the causal effect of diet on the risk of OCC. Six types of diets, including champagne or white wine (sample size: 175,549), red wine (sample size: 211,628), dried fruit (sample size: 409,125), fresh fruit (sample size: 433,186), average weekly beer plus cider (sample size: 327,634), and never eat eggs, dairy, wheat, sugar: wheat products (sample size: 461,046), were included as exposures. Summary statistics of OCC were obtained from a genome-wide association study (GWAS) including 1223 OCC cases and 2928 controls of European ancestry. We used the inverse-variance weighted (IVW) method as the primary method, with the weighted median and MR Egger used as supplementary methods for causal inference. Sensitivity analyses, including the Cochran Q test, Egger intercept test, MR-PRESSO test, and leave-one-out analysis, were performed to verify the robustness of the MR results. RESULTS The univariate MR showed that champagne or white wine consumption (β = 11.80, p = 0.006), average weekly beer plus cider intake (β = 4.73, p = 0.003), red wine consumption (β = 8.22, p = 0.024), and the never eat eggs, dairy, wheat, sugar: wheat products (β = 14.34, p = 0.002) increased the risk of OCC; Whereas fresh fruit consumption (β = - 3.20, p = 0.038), dried fruit consumption (β = - 2.98, p = 0.012) were protective. The multivariable MR estimates, after controlling for other diets, were as follows: when controlling for the never eat eggs, dairy, wheat, sugar: wheat products and fresh fruit consumption, the effect of average weekly beer plus cider intake on OCC risk was significant, and when controlling for dried fruit consumption, the effect had a P-value close to significant (p = 0.052). The two-step MR showed human characterization: When assessing the impact of dried fruit consumption and fresh fruit consumption, we observed a mediation effect of appendicular lean mass. Furthermore, when assessing the impact of dried fruit consumption and average weekly beer plus cider intake, we observed a mediation effect of standing height. Human system: When exploring the influence of never eat eggs, dairy, wheat, sugar: wheat products, we found a mediation effect of the occurrence of hypothyroidism. Likewise, when assessing the impact of fresh fruit consumption, we found the mediation effect of other diseases of the digestive system. Human metabolite: For the never eat eggs, dairy, wheat, sugar: wheat products, we observed a mediation effect of triglyceride levels in LDL and triglyceride levels in small LDL. CONCLUSIONS This study provides champagne or white wine consumption, average weekly beer plus cider intake, red wine consumption, and the never eat eggs, dairy, wheat, sugar: wheat products may increased the risk of OCC; Whereas fresh fruit and dried fruit consumption may were protective. This study analyzed the pathways by which dietary factors influence OCC in aspect of human characterization, human system, and human metabolite. These findings may help inform future research on preventing or treating OCC by modifying dietary factors.
Collapse
Affiliation(s)
- Shiyang Zhao
- Department of Stomatology, Jining Medical University, Shandong, 272067, China
| | - Xichen Liu
- Department of Stomatology, Jining Medical University, Shandong, 272067, China
| | - Qingsen Wang
- Department of Stomatology, Jining Medical University, Shandong, 272067, China
| | - Shudong Xiao
- Department of Stomatology, Jining Medical University, Shandong, 272067, China
| | - Wanting Wang
- Department of Stomatology, Jining Medical University, Shandong, 272067, China
| | - Xinyue Dong
- Department of Stomatology, Jining Medical University, Shandong, 272067, China
| | - Yourong Jiang
- Department of Stomatology, Jining Medical University, Shandong, 272067, China.
| | - Wen Wu
- Department of Stomatology, Jining Medical University, Shandong, 272067, China.
| |
Collapse
|
20
|
Tambets R, Kronberg J, van der Graaf A, Jesse M, Abner E, Võsa U, Rahu I, Taba N, Kolde A, Yarish D, Estonian Biobank Research Team, Fischer K, Kutalik Z, Esko T, Alasoo K, Palta P. Genome-wide association study for circulating metabolic traits in 619,372 individuals. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.10.15.24315557. [PMID: 40297438 PMCID: PMC12036396 DOI: 10.1101/2024.10.15.24315557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/30/2025]
Abstract
Interpreting genetic associations with complex traits can be greatly improved by detailed understanding of the molecular consequences of these variants. However, although genome-wide association studies (GWAS) for common complex diseases routinely profile 1M+ individuals, studies of molecular phenotypes have lagged behind. We performed a GWAS meta-analysis for 249 circulating metabolic traits in the Estonian Biobank and the UK Biobank in up to 619,372 individuals, identifying 88,604 significant locus-metabolite associations and 8,774 independent lead variants, including 987 lead variants with a minor allele frequency less than 1%. We demonstrate how common and low-frequency associations converge on shared genes and pathways, bridging the gap between rare-variant burden testing and common-variant GWAS. We used Mendelian randomisation (MR) to explore putative causal links between metabolic traits, coronary artery disease and type 2 diabetes (T2D). Surprisingly, up to 85% of the tested metabolite-disease pairs had statistically significant genome-wide MR estimates, likely reflecting complex indirect effects driven by horisontal pleiotropy. To avoid these pleiotropic effects, we used cis-MR to test the phenotypic impact of inhibiting specific drug targets. We found that although plasma levels of branched-chain amino acids (BCAAs) have been associated with T2D in both observational and genome-wide MR studies, inhibiting the BCAA catabolism pathway to lower BCAA levels is unlikely to reduce T2D risk. Our publicly available results provide a valuable novel resource for GWAS interpretation and drug target prioritisation.
Collapse
Affiliation(s)
- Ralf Tambets
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - Jaanika Kronberg
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | | | - Mihkel Jesse
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - Erik Abner
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Urmo Võsa
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Ida Rahu
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - Nele Taba
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Anastassia Kolde
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Institute of Mathematics and Statistics, University of Tartu, Tartu, Estonia
| | | | | | - Krista Fischer
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
| | - Zoltán Kutalik
- Department of Computational Biology, University of Lausanne, Lausanne, Switzerland
- University Center for Primary Care and Public Health, Unisanté, University of Lausanne, Lausanne, Switzerland
| | - Tõnu Esko
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Kaur Alasoo
- Institute of Computer Science, University of Tartu, Tartu, Estonia
| | - Priit Palta
- Estonian Genome Centre, Institute of Genomics, University of Tartu, Tartu, Estonia
| |
Collapse
|
21
|
Lin C, Xia M, Dai Y, Huang Q, Sun Z, Zhang G, Luo R, Peng Q, Li J, Wang X, Lin H, Gao X, Tang H, Shen X, Wang S, Jin L, Hao X, Zheng Y. Cross-ancestry analyses of Chinese and European populations reveal insights into the genetic architecture and disease implication of metabolites. CELL GENOMICS 2025; 5:100810. [PMID: 40118068 PMCID: PMC12008806 DOI: 10.1016/j.xgen.2025.100810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 01/22/2025] [Accepted: 02/17/2025] [Indexed: 03/23/2025]
Abstract
Differential susceptibilities to various diseases and corresponding metabolite variations have been documented across diverse ethnic populations, but the genetic determinants of these disparities remain unclear. Here, we performed large-scale genome-wide association studies of 171 directly quantifiable metabolites from a nuclear magnetic resonance-based metabolomics platform in 10,792 Han Chinese individuals. We identified 15 variant-metabolite associations, eight of which were successfully replicated in an independent Chinese population (n = 4,480). By cross-ancestry meta-analysis integrating 213,397 European individuals from the UK Biobank, we identified 228 additional variant-metabolite associations and improved fine-mapping precision. Moreover, two-sample Mendelian randomization analyses revealed evidence that genetically predicted levels of triglycerides in high-density lipoprotein were associated with a higher risk of coronary artery disease and that of glycine with a lower risk of heart failure in both ancestries. These findings enhance our understanding of the shared and specific genetic architecture of metabolites as well as their roles in complex diseases across populations.
Collapse
Affiliation(s)
- Chenhao Lin
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China; College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, Fujian 350002, China
| | - Mingfeng Xia
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yuxiang Dai
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Qingxia Huang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Zhonghan Sun
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Guoqing Zhang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China; National Genomics Data Center& Bio-Med Big Data Center, University of Chinese Academy of Sciences, Chinese Academy of Science, Shanghai 200031, China
| | - Ruijin Luo
- Shanghai Southgene Technology Co., Ltd., Shanghai 201203, China
| | - Qianqian Peng
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jinxi Li
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Xiaofeng Wang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China; Fudan University-the People's Hospital of Rugao Joint Research Institute of Longevity and Aging, Rugao, Jiangsu 226500, China
| | - Huandong Lin
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Xin Gao
- Department of Endocrinology and Metabolism, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Huiru Tang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Xia Shen
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China; Center for Intelligent Medicine Research, Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, Guangdong 511400, China
| | - Sijia Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China.
| | - Xingjie Hao
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China.
| | - Yan Zheng
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, Center for Evolutionary Biology, and School of Life Sciences, Fudan University, Shanghai 200433, China; Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
22
|
Wadström BN, Wulff AB, Pedersen KM, Nordestgaard BG. Small Remnants versus Large Triglyceride-Rich Lipoproteins in Risk of Atherosclerotic Cardiovascular Disease. Clin Chem 2025; 71:463-473. [PMID: 39882976 DOI: 10.1093/clinchem/hvae222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/18/2024] [Indexed: 01/31/2025]
Abstract
BACKGROUND Small remnants may penetrate the arterial intima more efficiently compared to large triglyceride-rich lipoproteins (TGRL). We tested the hypothesis that the importance of remnant cholesterol for the risk of atherosclerotic cardiovascular disease (ASCVD) may depend on the size of the remnants and TGRL carrying cholesterol. METHODS The cholesterol content of small remnants and large TGRL were measured in 25 572 individuals from the Copenhagen General Population Study (2003-2015) and in 222 721 individuals from the UK Biobank (2006-2010) using nuclear magnetic resonance spectroscopy. In the Copenhagen cohort during up to 15 years of follow-up and in the UK Biobank cohort during up to 16 years of follow-up, the numbers of individuals diagnosed with ASCVD (=myocardial infarction, ischemic stroke, and peripheral artery disease) in national health registries were 3869 and 11 424, respectively. RESULTS Compared to individuals with low cholesterol content in both small remnants and large TGRL (cutpoints were median cholesterol content), multivariable-adjusted hazard ratios for risk of ASCVD were 1.21 (95% confidence interval: 1.07-1.37) for individuals with high cholesterol content in small remnants only and 0.94 (0.83-1.07) for individuals with high cholesterol content in large TGRL only; the multivariable-adjusted hazard ratio for risk of ASCVD per 10 percentile-units higher cholesterol content in small remnants vs that in large TGRL was 1.04 (1.01-1.07). In the UK Biobank cohort, corresponding hazard ratios were 1.11 (1.03-1.20), 1.01 (0.93-1.09), and 1.05 (1.04-1.07), respectively. CONCLUSION The importance of remnant cholesterol for the risk of ASCVD may depend on the size of the TGRL and remnants carrying cholesterol.
Collapse
Affiliation(s)
- Benjamin N Wadström
- Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anders B Wulff
- Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kasper M Pedersen
- Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Børge G Nordestgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
23
|
Wadström BN, Borges MC, Wulff AB, Smith GD, Sanderson E, Nordestgaard BG. Elevated Remnant and LDL Cholesterol and the Risk of Peripheral Artery Disease: A Mendelian Randomization Study. J Am Coll Cardiol 2025; 85:1353-1368. [PMID: 40139892 DOI: 10.1016/j.jacc.2024.12.033] [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: 10/16/2024] [Revised: 12/16/2024] [Accepted: 12/21/2024] [Indexed: 03/29/2025]
Abstract
BACKGROUND Elevated remnant cholesterol and low-density lipoprotein (LDL) cholesterol both increase the risk of coronary artery disease (CAD), but it is not known if the same is true for peripheral artery disease (PAD). OBJECTIVES This study tested the hypothesis that elevated remnant cholesterol and LDL cholesterol, each independent of the other, have causal effects on risk of PAD. METHODS The authors constructed genetic scores from variants near genes known to directly affect levels of remnant cholesterol and LDL cholesterol, identified through a genome-wide association study of individuals in the UK Biobank. Univariable (remnant cholesterol and LDL cholesterol genetic scores separately) and multivariable (remnant cholesterol and LDL cholesterol genetic scores combined) Mendelian randomization were used to estimate the causal effects of higher remnant cholesterol and LDL cholesterol levels on ORs for PAD (n = 38,414 cases and 758,308 controls) and CAD (n = 221,445 cases and 770,615 controls). RESULTS Increments in remnant and LDL genetic scores corresponding to 1 mmol/L (39 mg/dL) higher remnant and LDL cholesterol, respectively, were associated with univariable ORs for PAD of 2.72 (95% CI: 2.10-3.52) and 1.37 (95% CI: 1.25-1.51); corresponding multivariable ORs were 2.16 (95% CI: 1.49-3.12) and 1.14 (95% CI: 1.00-1.30). For CAD, corresponding univariable ORs were 2.92 (95% CI: 2.34-3.64) and 1.67 (95% CI: 1.56-1.79), whereas multivariable ORs were 1.86 (95% CI: 1.39-2.47) and 1.44 (95% CI: 1.29-1.60). Scaled to 1 SD increments in remnant cholesterol and LDL cholesterol, corresponding univariable ORs were 1.37 (95% CI: 1.27-1.49) and 1.29 (95% CI: 1.20-1.39) for PAD, and 1.40 (95% CI: 1.31-1.51) and 1.51 (95% CI: 1.43-1.59) for CAD; corresponding multivariable ORs were 1.28 (95% CI: 1.14-1.43) and 1.11 (95% CI: 1.00-1.23) for PAD, and 1.22 (95% CI: 1.11-1.33) and 1.34 (95% CI: 1.23-1.46) for CAD. CONCLUSIONS Elevated remnant cholesterol had a causal effect on risk of PAD even after accounting for elevated LDL cholesterol, whereas most of the causal effect of elevated LDL cholesterol on risk of PAD was dependent on simultaneously elevated remnant cholesterol. These results indicate that remnant cholesterol may be the major cholesterol fraction responsible for increased risk of PAD. Future studies should investigate the biological mechanisms behind these findings to find improved therapies for prevention and treatment of PAD.
Collapse
Affiliation(s)
- Benjamin Nilsson Wadström
- Medical Research Council (MRC) Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom; Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark; The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Maria Carolina Borges
- Medical Research Council (MRC) Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Anders Berg Wulff
- Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark; The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - George Davey Smith
- Medical Research Council (MRC) Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom; National Institute for Health Research (NIHR), Biomedical Research Centre, University of Bristol, Bristol, United Kingdom
| | - Eleanor Sanderson
- Medical Research Council (MRC) Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom; Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Børge Grønne Nordestgaard
- Department of Clinical Biochemistry, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark; The Copenhagen General Population Study, Copenhagen University Hospital-Herlev and Gentofte, Herlev, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
24
|
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
|
25
|
Venkataraghavan S, Pankow JS, Boerwinkle E, Fornage M, Selvin E, Ray D. Epigenome-wide association study of incident type 2 diabetes in Black and White participants from the Atherosclerosis Risk in Communities Study. Diabetologia 2025; 68:815-834. [PMID: 39971753 PMCID: PMC12054846 DOI: 10.1007/s00125-024-06352-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 05/29/2024] [Indexed: 02/21/2025]
Abstract
AIMS/HYPOTHESIS DNA methylation studies of incident type 2 diabetes in US populations are limited and to our knowledge none include individuals of African descent. We aimed to fill this gap by identifying methylation sites (CpG sites) and regions likely influencing the development of type 2 diabetes using data from Black and White individuals from the USA. METHODS We prospectively followed 2091 Black and 1029 White individuals without type 2 diabetes from the Atherosclerosis Risk in Communities study over a median follow-up period of 17 years, and performed an epigenome-wide association analysis of blood-based methylation levels with incident type 2 diabetes using Cox regression. We assessed whether significant CpG sites were associated with incident type 2 diabetes independently of BMI or fasting glucose at baseline. We estimated variation in incident type 2 diabetes accounted for by the major non-genetic risk factors and the significant CpG sites. We also examined groups of methylation sites that were differentially methylated. We performed replication of previously discovered CpG sites associated with prevalent and/or incident type 2 diabetes. All analyses were adjusted for batch effects, cell-type proportions and relevant confounders. RESULTS At an epigenome-wide threshold (10-7), we detected seven novel diabetes-associated CpG sites, of which the sites at MICOS10 (cg05380846: HR 0.89, p=8.4 × 10-12), ZNF2 (cg01585592: HR 0.88, p=1.6 × 10-9), JPH3 (cg16696007: HR 0.87, p=7.8 × 10-9) and GPX6 (cg02793507: HR 0.85, p=2.7 × 10-8; cg00647063: HR 1.20, p=2.5 × 10-8) were identified in Black adults; chr17q25 (cg16865890: HR 0.8, p=6.9 × 10-8) in White adults; and chr11p15 (cg13738793: HR 1.11, p=7.7 × 10-8) in the meta-analysed group. The JPH3 and GPX6 sites remained epigenome-wide significant on adjustment for BMI, while only the JPH3 site retained significance after adjusting for fasting glucose. We replicated known type 2 diabetes-associated CpG sites, including cg19693031 at TXNIP, cg00574958 at CPT1A, cg16567056 at PLCB2, cg11024682 at SREBF1, cg08857797 at VPS25 and cg06500161 at ABCG1, three of which were replicated in Black adults at the epigenome-wide threshold and all of which had directionally consistent effects. We observed a modest increase in type 2 diabetes variance explained by the significantly associated CpG sites over and above traditional type 2 diabetes risk factors and fasting glucose (26.2% vs 30.5% in Black adults; 36.9% vs 39.4% in White adults). At the Šidák-corrected significance threshold of 5%, our differentially methylated region (DMR) analyses revealed several clusters of significant CpG sites, including a DMR consisting of a previously discovered CpG site at ADCY7 (pBlack=1.8 × 10-4, pWhite=3.6 × 10-3, pAll=1.6 × 10-9) and a DMR consisting of the promoter region of TP63 (pBlack=7.4 × 10-4, pWhite=3.9 × 10-3, pAll=1.4 × 10-5), which were differentially methylated across all racial and ethnic groups. CONCLUSIONS/INTERPRETATION This study illustrates improved discovery of CpG sites and regions by leveraging both individual CpG site analysis and DMR analyses in an unexplored population. Our findings include genes linked to diabetes in experimental studies (e.g. GPX6, JPH3 and TP63). The JPH3 and GPX6 sites were likely associated with incident type 2 diabetes independently of BMI. All the CpG sites except that at JPH3 were likely consequences of elevated glucose. Replication in African-descent individuals of CpG sites previously discovered mostly in individuals of European descent indicates that some of these methylation-type 2 diabetes associations are robust across racial and ethnic groups. This study is a first step towards understanding the influence of methylation on the incidence of type 2 diabetes and its disparity in two major racial and ethnic groups in the USA. It paves the way for future studies to investigate causal relationships between type 2 diabetes and the CpG sites and potentially elucidate molecular targets for intervention.
Collapse
Affiliation(s)
- Sowmya Venkataraghavan
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - James S Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Eric Boerwinkle
- The University of Texas Health School of Public Health, Houston, TX, USA
| | - Myriam Fornage
- Brown Foundation Institute for Molecular Medicine, The University of Texas Health Science Center, Houston, TX, USA
| | - Elizabeth Selvin
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
- Welch Center for Prevention, Epidemiology, & Clinical Research, Johns Hopkins University, Baltimore, MD, USA
| | - Debashree Ray
- Department of Epidemiology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
- Department of Biostatistics, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
26
|
Schneider KM, Cao F, Huang HYR, Chen L, Chen Y, Gong R, Raptis A, Creasy KT, Clusmann J, van Haag F, Koop P, Guillot A, Luedde T, Loomba R, Francque S, Schneider CV. The Lipidomic Profile Discriminates Between MASLD and MetALD. Aliment Pharmacol Ther 2025; 61:1357-1371. [PMID: 39935287 PMCID: PMC11950808 DOI: 10.1111/apt.70012] [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: 10/06/2024] [Revised: 10/22/2024] [Accepted: 01/29/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND The recent consensus statement redefined steatotic liver diseases. Metabolic dysfunction-associated steatotic liver disease (MASLD) and metabolic dysfunction and alcohol-related liver disease (MetALD) now represent distinct disease entities. However, biomarkers that differentiate MASLD and MetALD remain largely unknown. AIMS To identify lipidomic biomarkers with discriminatory potential for distinguishing MetALD from MASLD. METHODS Using the UK Biobank dataset, 40,534 people with available MRI liver scans were analysed. A total of, 11,217 cases with a proton density fat fraction (PDFF) ≥ 5% were identified as having steatotic liver disease. Among these, lipidomic profiles were obtained for 5539 MASLD and 462 MetALD cases. A total of, 250 plasma lipidomic and metabolomic parameters were analysed. Mendelian randomisation (MR) analysis was used to confirm the association between alcohol consumption and the lipidomic biomarkers. RESULTS When comparing the top 30 differentially expressed lipidomic biomarkers predicting MetALD compared to MASLD, the majority were related to HDL and were significantly overrepresented at both analysed time points. The top five metabolites were: acetoacetate, 3-hydroxybutyrate, phospholipids in Large HDL, concentration of large HDL particles, free cholesterol in large HDL. The sensitivity analysis comparing alcohol-related liver disease to MASLD revealed similar associations, suggesting that the HDL signature is stable over time. Additionally, MR analysis further confirmed that alcohol consumption was associated with increased levels of HDL-related metabolites. CONCLUSION Our findings indicate that HDL-centric lipidomic markers, particularly those within the larger and medium HDL subfraction, may differentiate MetALD from MASLD. Further longitudinal and experimental studies are warranted to validate these findings and assess their clinical implications.
Collapse
Affiliation(s)
- Kai Markus Schneider
- Department of Internal Medicine III, Gastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
- Department of Medicine I, Deptartment of Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Carl Gustav CarusTUD Dresden University of TechnologyDresdenGermany
- Center for Regenerative Therapies Dresden (CRTD)Technische Universität (TU) DresdenDresdenGermany
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav CarusTUD Dresden University of TechnologyDresdenGermany
| | - Feng Cao
- Department of Internal Medicine III, Gastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
| | - Helen Ye Rim Huang
- Department of Internal Medicine III, Gastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
| | - Lanlan Chen
- Department of Hepatology & Gastroenterology, Campus Virchow‐Klinikum and Campus Charité MitteCharité – Universitätsmedizin BerlinBerlinGermany
| | - Yazhou Chen
- Department of Internal Medicine III, Gastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
| | - Rongpeng Gong
- Department of Internal Medicine III, Gastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
| | - Anastasia Raptis
- Department of Internal Medicine III, Gastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
| | - Kate Townsend Creasy
- Department of Biobehavioral Health Sciences, School of NursingUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Jan Clusmann
- Department of Internal Medicine III, Gastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav CarusTUD Dresden University of TechnologyDresdenGermany
| | - Felix van Haag
- Department of Internal Medicine III, Gastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
| | - Paul‐Henry Koop
- Department of Internal Medicine III, Gastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
| | - Adrien Guillot
- Department of Hepatology & Gastroenterology, Campus Virchow‐Klinikum and Campus Charité MitteCharité – Universitätsmedizin BerlinBerlinGermany
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical FacultyHeinrich‐Heine‐UniversityDüsseldorfGermany
| | - Rohit Loomba
- MASLD Research Center, Division of Gastroenterology and Hepatology, Department of MedicineUniversity of California at San DiegoSan DiegoCaliforniaUSA
| | - Sven Francque
- Department of Gastroenterology HepatologyAntwerp University HospitalEdegemBelgium
- InflaMed Centre of Excellence, Laboratory for Experimental Medicine and Paediatrics, Translational Sciences in Inflammation and Immunology, Faculty of Medicine and Health SciencesUniversity of AntwerpWilrijkBelgium
| | - Carolin Victoria Schneider
- Department of Internal Medicine III, Gastroenterology, Metabolic Diseases and Intensive CareUniversity Hospital RWTH AachenAachenGermany
- The Institute for Translational Medicine and Therapeutics, the Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
27
|
Wang B, Yang Y, Yin Z, Yang W. The causal impact of body mass index on metabolic biomarkers and nonalcoholic fatty liver disease risk. Sci Rep 2025; 15:10314. [PMID: 40133380 PMCID: PMC11937590 DOI: 10.1038/s41598-024-84165-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 12/20/2024] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is a growing global health concern linked to obesity. METHODS This study employed a Mendelian randomization approach to explore the causal influence of BMI on metabolic biomarkers and the subsequent risk of NAFLD. We analyzed data from multiple sources, including 249 metabolic traits, to establish direct and mediating relationships among BMI, metabolic factors, and NAFLD risk. RESULTS Our findings revealed a significant positive correlation between BMI and NAFLD across various datasets. We identified 176 metabolites associated with BMI, of which 106 were also linked to NAFLD. Importantly, 86 metabolites were found to mediate the relationship between BMI and NAFLD risk. Specifically, elevated levels of branched-chain amino acids, triglycerides, and certain cholesterol esters were notably associated with increased NAFLD risk, whereas changes in free cholesterol and phospholipid levels also played critical roles. CONCLUSION This study highlights the complex interactions between BMI, metabolic biomarkers, and NAFLD risk. By elucidating these relationships, we highlight potential targets for interventions aimed at reducing NAFLD incidence in populations with elevated BMI, ultimately contributing to improved metabolic health.
Collapse
Affiliation(s)
- Bo Wang
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Yanjiang Yang
- Department of Rheumatology and Immunology, The People's Hospital of Qiandongnan Autonomous Prefecture, Kaili, 556000, Guizhou Province, China
| | - Zhaoqiang Yin
- Department of Minimally Invasive and Biliary Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China.
| | - Wenwen Yang
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, Gansu Province, China.
| |
Collapse
|
28
|
Chopra A, Song J, Weiner 3rd J, Beule D, Schaefer AS. Genetic analysis of cis-enhancers associated with bone mineral density and periodontitis in the gene SOST. PLoS One 2025; 20:e0319259. [PMID: 40127057 PMCID: PMC11932464 DOI: 10.1371/journal.pone.0319259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 01/27/2025] [Indexed: 03/26/2025] Open
Abstract
A haplotype block at the sclerostin (SOST) gene correlates with bone mineral density (BMD) and increased periodontitis risk in smokers. Investigating the putative causal variants within this block, our study aimed to elucidate the impact of linked enhancer elements on gene expression and to evaluate their role in transcription factor (TF) binding. Using CRISPR/dCas9 activation (CRISPRa) screening in SaOS-2 cells, we quantified disease-related enhancer activities regulating SOST expression. Additionally, in SaOS-2 cells, we investigated the influence of the candidate TFs CCAAT/enhancer-binding protein beta (CEBPB) on gene expression by antisense (GapmeR) knockdown, followed by RNA sequencing. The periodontitis-linked SNP rs9783823 displayed a significant cis-activating effect (25-fold change in SOST expression), with the C-allele containing a CEBPB binding motif (position weight matrix (PWM) = 0.98, Pcorrected = 7.7 x 10-7). CEBPB knockdown induced genome-wide upregulation but decreased epithelial-mesenchymal transition genes (P = 0.71, AUC = 2.2 x 10-11). This study identifies a robust SOST cis-activating element linked to BMD and periodontitis, carrying CEBPB binding sites, and highlights CEBPB's impact on epithelial-mesenchymal transition.
Collapse
Affiliation(s)
- Avneesh Chopra
- Department of Periodontology, Oral Medicine and Oral Surgery, Institute for Dental and Craniofacial Sciences, Charité-University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jiahui Song
- Department of Periodontology, Oral Medicine and Oral Surgery, Institute for Dental and Craniofacial Sciences, Charité-University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | | | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health, Berlin, Germany
| | - Arne S. Schaefer
- Department of Periodontology, Oral Medicine and Oral Surgery, Institute for Dental and Craniofacial Sciences, Charité-University Medicine Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
29
|
Zhang H, Zhou Z, Gu J, Lin Y, Yan Y, Chen X, Fan M, Huang Y. Genetic insights of lipid metabolism and lipid-lowering drugs with Lewy body dementia risk: Evidence from Mendelian randomization. Prog Neuropsychopharmacol Biol Psychiatry 2025; 137:111282. [PMID: 39929371 DOI: 10.1016/j.pnpbp.2025.111282] [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: 10/24/2024] [Revised: 01/29/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND Lewy body dementia (LBD) is the second common dementia, with unclear mechanisms and limited treatment options. Dyslipidemia has been implicated in LBD, but the role of lipid-lowering drugs remains underexplored. This study aims to investigate the association between lipid traits, drug targets, and LBD risk using Mendelian Randomization (MR) analysis. METHODS We performed univariable and multivariable MR analyses to evaluate the causal effects of lipid traits on the risk of LBD. Then, drug-target MR analysis and subtype analysis were conducted to evaluate the effects of lipid-lowering therapies on LBD. RESULTS In univariable MR, genetically predicted low-density lipoprotein cholesterol (LDL-C) and remnant cholesterol (RC) levels were associated with an increased risk of LBD. Mediation analysis suggested a potential interaction between LDL-C and RC in influencing LBD risk. Drug-target MR analysis identified significant associations between genetically proxied inhibition of ANGPTL3, CETP, and HMGCR and LBD risk. CONCLUSION This MR analysis provided evidence that elevated LDL-C and RC may increase the risk of LBD. Additionally, targeting ANGPTL3, CETP, and HMGCR may represent potential therapeutic strategies for the prevention or treatment of LBD.
Collapse
Affiliation(s)
- Hanyu Zhang
- Department of General Medicine, Huashan Hospital, Fudan University, Shanghai, PR China
| | - Zengyuan Zhou
- Department of Nutrition, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Onclogy, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jie Gu
- Department of General Medicine, Huashan Hospital, Fudan University, Shanghai, PR China
| | - Yingnan Lin
- Department of General Medicine, Huashan Hospital, Fudan University, Shanghai, PR China
| | - Yunyun Yan
- Department of General Medicine, Huashan Hospital, Fudan University, Shanghai, PR China
| | - Xiaonan Chen
- Department of General Medicine, Huashan Hospital, Fudan University, Shanghai, PR China
| | - Meixiang Fan
- Department of General Medicine, Huashan Hospital, Fudan University, Shanghai, PR China
| | - Yanyan Huang
- Department of General Medicine, Huashan Hospital, Fudan University, Shanghai, PR China; National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan Universiy, Shanghai, PR China; Tianqiao and Chrissy Chen Institute Clinic Translational Research Center, Shanghai, PR China; Department of Geriatrics, Huashan Hospital, Fudan University, Shanghai, PR China.
| |
Collapse
|
30
|
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
|
31
|
Jiang D, Yang W, Zhang Y. Fatty acids and colorectal cancer: Insights from Mendelian randomization. Medicine (Baltimore) 2025; 104:e41768. [PMID: 40101081 PMCID: PMC11922435 DOI: 10.1097/md.0000000000041768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 02/17/2025] [Indexed: 03/20/2025] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers worldwide, necessitating the identification of risk factors and preventive measures. Fatty acids, vital nutrients involved in various bodily functions, have been linked to CRC; however, findings are inconsistent. This Mendelian randomization study utilized data from the UK Biobank and included 18 fatty acid-related phenotypes. We used single-nucleotide polymorphisms as instrumental variables to examine the Causal connections between fatty acids and CRC. Statistical analysis involved the inverse-variance-weighted, Mendelian randomization-Egger, and weighted median methods to ensure robust findings. Our analysis revealed that docosahexaenoic acid and omega-3 fatty acids were positively associated with CRC risk. No significant associations were found between CRC and total fatty acids, saturated fatty acids, polyunsaturated fatty acids, or monounsaturated fatty acids. The degree of unsaturation was positively associated with CRC, while the ratio of omega-6 to omega-3 fatty acids was negatively associated. The study highlights a positive association between docosahexaenoic acid, omega-3 fatty acids, and CRC, suggesting that specific fatty acids may influence CRC risk. Further research in diverse populations is needed to confirm these findings and explore the underlying mechanisms.
Collapse
Affiliation(s)
- Dengge Jiang
- Xi’AN No.1 Hospital, The First Affiliated Hospital of Northwest University, Xi’an, Shaanxi, China
| | - Wenwen Yang
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu, China
| | - Yu Zhang
- Xi’AN No.1 Hospital, The First Affiliated Hospital of Northwest University, Xi’an, Shaanxi, China
| |
Collapse
|
32
|
Xia X, Tie X, Hong M, Yin W. Exploration of the causal relationship and mechanisms between serum albumin and venous thrombosis: a bidirectional mendelian randomization analysis and bioinformatics study. Thromb J 2025; 23:17. [PMID: 40033322 DOI: 10.1186/s12959-025-00700-4] [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: 10/08/2024] [Accepted: 02/11/2025] [Indexed: 03/05/2025] Open
Abstract
BACKGROUND To explore the causal relationship between serum albumin and venous thromboembolism (VTE) comprises deep vein thrombosis (DVT) and its consequential condition, pulmonary embolism (PE), through Mendelian randomization (MR) design, seeking to clarify the protective roles of albumin in the development of venous thrombosis. METHODS We performed a bidirectional two-sample Mendelian randomization analysis utilizing albumin genome-wide association study (GWAS) data alongside VTE datasets from various sources. Additionally, to minimize heterogeneity across different datasets, a meta-analysis of the Mendelian randomization results was conducted. Furthermore, genes associated with such exposures were identified to unravel how exposure impacts outcomes. This was followed by applying bioinformatics techniques for gene enrichment analysis and employing the Cytoscape software to pinpoint the hub genes. RESULTS The findings from the meta-analysis of the Mendelian randomization indicate that reduced levels of albumin are associated with an elevated risk of VTE (OR = 0.739, 95% CI: 0.695 to 0.787, P = 1.82e-9), DVT (OR = 0.700, 95% CI: 0.646 to 0.772, P = 2.96e-15), and PE (OR = 0.717, 95% CI: 0.647 to 0.793, P = 1.74e-10). Bioinformatics analysis revealed that serum albumin primarily protects against VTE by influencing inflammation and cytokines. CONCLUSIONS Our bidirectional MR analysis confirmed a substantial causal association linking serum albumin to VTE. Bioinformatics analysis revealed that this causal link is mediated by the immune response through inflammation and cytokines.
Collapse
Affiliation(s)
- Xuemei Xia
- Department of Critical Care Medicine, West China Hospital, Sichuan University, No.37, Guoxue Alley, Wuhou District, Chengdu, Sichuan Province, China
| | - Xin Tie
- Department of Critical Care Medicine, West China Hospital, Sichuan University, No.37, Guoxue Alley, Wuhou District, Chengdu, Sichuan Province, China
| | - Maolin Hong
- Department of Critical Care Medicine, West China Hospital, Sichuan University, No.37, Guoxue Alley, Wuhou District, Chengdu, Sichuan Province, China
| | - Wanhong Yin
- Department of Critical Care Medicine, West China Hospital, Sichuan University, No.37, Guoxue Alley, Wuhou District, Chengdu, Sichuan Province, China.
| |
Collapse
|
33
|
Yang Y, Wang X, Yang W. Dried fruit, acetate, and asthma: a mediation Mendelian randomization analysis. J Asthma 2025; 62:410-415. [PMID: 39311580 DOI: 10.1080/02770903.2024.2408284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/15/2024] [Accepted: 09/20/2024] [Indexed: 10/02/2024]
Abstract
BACKGROUND Asthma is a common chronic condition with increasing prevalence. Diet, including dried fruit consumption, has been linked to asthma risk, but the mechanisms remain unclear. This study investigates how dried fruit consumption affects asthma risk, focusing on acetate as a potential mediator. METHODS We used Mendelian Randomization (MR) to analyze the relationships between dried fruit intake, acetate levels, and asthma. We applied three MR methods-Inverse-Variance Weighted (IVW), Weighted Median (WM), and MR-Egger-to determine causal effects. RESULTS Dried fruit intake was inversely associated with asthma risk (IVW: β = -0.506, p = 0.0135) and positively associated with acetate levels (IVW: β = 0.269, p < 0.0001). Higher acetate levels were also linked to lower asthma risk (IVW: β=-0.361, p < 0.0001). Mediation analysis showed that acetate mediates approximately 19.22% of the effect of dried fruit on asthma risk. CONCLUSION Dried fruit consumption reduces asthma risk, partly through increasing acetate levels. This acetate-mediated pathway accounts for about 20% of the effect, suggesting potential for dietary strategies in asthma prevention and management. Further research could enhance the understanding and applicability of these findings.
Collapse
Affiliation(s)
- Yanjiang Yang
- Department of Rheumatology and Immunology, The people's Hospital of Qiandongnan Autonomous Prefecture, Kaili, Guizhou Province, China
| | - Xiaorui Wang
- School of Pharmacy, Chengdu University, Chengdu, China
| | - Wenwen Yang
- The First Clinical Medical College, Lanzhou University, Lanzhou, Gansu Province, China
| |
Collapse
|
34
|
Hu Y, Cui X, Lu M, Guan X, Li Y, Zhang L, Lin L, Zhang Z, Zhang M, Hao J, Wang X, Huan J, Li Y, Li C. Body Fat Distribution and Ectopic Fat Accumulation as Mediator of Diabetogenic Action of Lipid-Modifying Drugs: A Mediation Mendelian Randomization Study. Mayo Clin Proc 2025; 100:424-439. [PMID: 39918451 DOI: 10.1016/j.mayocp.2024.10.018] [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: 04/24/2024] [Revised: 10/06/2024] [Accepted: 10/25/2024] [Indexed: 05/08/2025]
Abstract
OBJECTIVE To investigate the causal relationship between various lipid-modifying drugs and new-onset diabetes, as well as the mediators contributing to this relationship. METHODS Mediation Mendelian randomization was performed to investigate the causal effect of lipid-modifying drug targets on type 2 diabetes (T2D) outcomes and the proportion of this association that is mediated through ectopic fat accumulation traits. Specific sets of variants in or near genes that encode 11 lipid-modifying drug targets (LDLR, HMGCR, NPC1L1, PCSK9, APOB, ABCG5/ABCG8, LPL, PPARA, ANGPTL3, APOC3, and CETP; for expansion of gene symbols, use search tool at www.genenames.org) were extracted. Random effects inverse variance weighted were performed to evaluate the causal effects among outcomes. Mediation analyses were performed to identify the mediators of the association between lipid-modifying drugs and T2D. The study was conducted from November 10, 2023, to April 2, 2024 RESULTS: The genetic mimicry of HMGCR and APOB inhibition was associated with an increased T2D risk, whereas the genetic mimicry of LPL enhancement was linked to a lower T2D risk. Gluteofemoral adipose tissue volume was a mediator for explaining 9.52% (P=.002), 16.90% (P=.03), and 10.50% (P=.003) of the total effect of HMGCR, APOB, and LPL on T2D susceptibility, respectively. Liver fat was a mediator for explaining 21.12% (P=.005), 12.28% (P=.03), and 9.84% (P=.005) of the total effect of HMGCR, APOB, and LPL on T2D susceptibility, respectively. CONCLUSION Our findings support the hypothesis that liver fat and gluteofemoral adipose tissue play a mediating role in the prodiabetic effects of HMGCR and APOB inhibition, as well as in the antidiabetic effects of LPL enhancement.
Collapse
Affiliation(s)
- Yuanlong Hu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xinhai Cui
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Mengkai Lu
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiuya Guan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yuan Li
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Lei Zhang
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Lin Lin
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Zhiyuan Zhang
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Muxin Zhang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Jiaqi Hao
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiaojie Wang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macau, China
| | - Jiaming Huan
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Yunlun Li
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China; Department of Cardiovascular, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
| | - Chao Li
- Innovation Research Institute of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China.
| |
Collapse
|
35
|
Qiu Y, Song B, Yin Z, Wang M, Tao Y, Xie M, Duan A, Chen Z, Si K, Wang Z. Novel insights into causal effects of serum lipids, lipid metabolites, and lipid-modifying targets on the risk of intracerebral aneurysm. Eur Stroke J 2025; 10:236-247. [PMID: 39081035 PMCID: PMC11569451 DOI: 10.1177/23969873241265019] [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: 03/31/2024] [Accepted: 06/05/2024] [Indexed: 11/19/2024] Open
Abstract
INTRODUCTION Different serum lipid and lipid-lowering agents are reported to be related to the occurrence of intracerebral aneurysm (IA). However, the causal relationship between them requires further investigation. PATIENTS AND METHODS Mendelian randomization (MR) analysis was performed on IA and its subtypes by using instrumental variants associated with six serum lipids, 249 lipid metabolic traits, and 10 lipid-lowering agents that were extracted from the largest genome-wide association study. Phenome-wide MR analyses were conducted to identify potential phenotypes associated with significant lipid-lowering agents. RESULTS After multiple comparison adjustments (p < 0.0083), genetically proxied triglyceride (TG) (odds ratio [OR] 1.25, 95% confidence interval [CI] 1.07-1.47, p = 0.005) and high-density lipoprotein cholesterol (HDL-C) levels (OR 0.93, 95% CI 0.89-0.98, p = 0.008) showed causal relationships with the risk of IA. Four lipid metabolic traits showed a causal relationship with the risk of IA (p < 0.0002). As confirmed by drug target MR, the causal relationship between the HMGCR target and IA, HMGCR target and subarachnoid hemorrhage (SAH), ANGPTL3 target and SAH, CETP target, and SAH remained statistically significant after multiple adjustments (p < 0.005). Additionally, phenome-wide MR did not identify other diseases linked to the significant lipid-lowering agent (p < 6.39 × 10-5). DISCUSSION AND CONCLUSION This study not only supports that serum lipids (TG and HDL-C) are associated with IA but also confirms the positive effect and absence of safety concerns of intervening HMGCR, ANGPTL3, and CETP targets in IA and its subtypes, opening new avenues for IA treatment.
Collapse
Affiliation(s)
- Youjia Qiu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Bingyi Song
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Ziqian Yin
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Menghan Wang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Yuchen Tao
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Minjia Xie
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Aojie Duan
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhouqing Chen
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Ke Si
- Department of Cardiac Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zhong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
36
|
Le MHN, Nguyen PK, Nguyen TPT, Nguyen HQ, Tam DNH, Huynh HH, Huynh PK, Le NQK. An in-depth review of AI-powered advancements in cancer drug discovery. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167680. [PMID: 39837431 DOI: 10.1016/j.bbadis.2025.167680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/12/2025] [Accepted: 01/16/2025] [Indexed: 01/23/2025]
Abstract
The convergence of artificial intelligence (AI) and genomics is redefining cancer drug discovery by facilitating the development of personalized and effective therapies. This review examines the transformative role of AI technologies, including deep learning and advanced data analytics, in accelerating key stages of the drug discovery process: target identification, drug design, clinical trial optimization, and drug response prediction. Cutting-edge tools such as DrugnomeAI and PandaOmics have made substantial contributions to therapeutic target identification, while AI's predictive capabilities are driving personalized treatment strategies. Additionally, advancements like AlphaFold highlight AI's capacity to address intricate challenges in drug development. However, the field faces significant challenges, including the management of large-scale genomic datasets and ethical concerns surrounding AI deployment in healthcare. This review underscores the promise of data-centric AI approaches and emphasizes the necessity of continued innovation and interdisciplinary collaboration. Together, AI and genomics are charting a path toward more precise, efficient, and transformative cancer therapeutics.
Collapse
Affiliation(s)
- Minh Huu Nhat Le
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; AIBioMed Research Group, Taipei Medical University, Taipei 110, Taiwan
| | - Phat Ky Nguyen
- International Master/Ph.D. Program in Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; AIBioMed Research Group, Taipei Medical University, Taipei 110, Taiwan.
| | | | - Hien Quang Nguyen
- Cardiovascular Research Department, Methodist Hospital, Merrillville, IN 46410, USA
| | - Dao Ngoc Hien Tam
- Regulatory Affairs Department, Asia Shine Trading & Service Co. LTD, Viet Nam
| | - Han Hong Huynh
- International Master Program for Translational Science, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Phat Kim Huynh
- Department of Industrial and Systems Engineering, North Carolina A&T State University, Greensboro, NC 27411, USA.
| | - Nguyen Quoc Khanh Le
- AIBioMed Research Group, Taipei Medical University, Taipei 110, Taiwan; In-Service Master Program in Artificial Intelligence in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan; Translational Imaging Research Center, Taipei Medical University Hospital, Taipei 110, Taiwan.
| |
Collapse
|
37
|
Wang Z, Chen S, Zhang F, Akhmedov S, Weng J, Xu S. Prioritization of Lipid Metabolism Targets for the Diagnosis and Treatment of Cardiovascular Diseases. RESEARCH (WASHINGTON, D.C.) 2025; 8:0618. [PMID: 39975574 PMCID: PMC11836198 DOI: 10.34133/research.0618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/15/2025] [Accepted: 01/29/2025] [Indexed: 02/21/2025]
Abstract
Background: Cardiovascular diseases (CVD) are a major global health issue strongly associated with altered lipid metabolism. However, lipid metabolism-related pharmacological targets remain limited, leaving the therapeutic challenge of residual lipid-associated cardiovascular risk. The purpose of this study is to identify potentially novel lipid metabolism-related genes by systematic genomic and phenomics analysis, with an aim to discovering potentially new therapeutic targets and diagnosis biomarkers for CVD. Methods: In this study, we conducted a comprehensive and multidimensional evaluation of 881 lipid metabolism-related genes. Using genome-wide association study (GWAS)-based mendelian randomization (MR) causal inference methods, we screened for genes causally linked to the occurrence and development of CVD. Further validation was performed through colocalization analysis in 2 independent cohorts. Then, we employed reverse screening using phenonome-wide association studies (PheWAS) and a drug target-drug association analysis. Finally, we integrated serum proteomic data to develop a machine learning model comprising 5 proteins for disease prediction. Results: Our initial screening yielded 54 genes causally linked to CVD. Colocalization analysis in validation cohorts prioritized this to 29 genes marked correlated with CVD. Comparison and interaction analysis identified 13 therapeutic targets with potential for treating CVD and its complications. A machine learning model incorporating 5 proteins for CVD prediction achieved a high accuracy of 96.1%, suggesting its potential as a diagnostic tool in clinical practice. Conclusion: This study comprehensively reveals the complex relationship between lipid metabolism regulatory targets and CVD. Our findings provide new insights into the pathogenesis of CVD and identify potential therapeutic targets and drugs for its treatment. Additionally, the machine learning model developed in this study offers a promising tool for the diagnosis and prediction of CVD, paving the way for future research and clinical applications.
Collapse
Affiliation(s)
- Zhihua Wang
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei 230001, China
- Institute of Endocrine and Metabolic Diseases, University of Science and Technology of China, Hefei 230001, China
| | - Shuo Chen
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei 230001, China
| | - Fanshun Zhang
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei 230001, China
| | - Shamil Akhmedov
- Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634012, Russia
| | - Jianping Weng
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei 230001, China
- Institute of Endocrine and Metabolic Diseases, University of Science and Technology of China, Hefei 230001, China
- Anhui Provincial Key Laboratory of Metabolic Health and Panvascular Diseases, Hefei 230001, China
| | - Suowen Xu
- Department of Endocrinology, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei 230001, China
- Institute of Endocrine and Metabolic Diseases, University of Science and Technology of China, Hefei 230001, China
- Anhui Provincial Key Laboratory of Metabolic Health and Panvascular Diseases, Hefei 230001, China
| |
Collapse
|
38
|
Sheng Q, Ma Y, Geng B, Chen J, Cheng J, Liu S, Li R, Li X, Wang J, Lu H, Gao F, Gao F. Serum amino acid alterations in hyperuricemia: potential targets for renal disease prevention. Amino Acids 2025; 57:16. [PMID: 39966264 PMCID: PMC11836093 DOI: 10.1007/s00726-025-03444-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 01/30/2025] [Indexed: 02/20/2025]
Abstract
Observational studies have linked uric acid (UA) levels and kidney disease to amino acid homeostasis, but the causal relationship is unclear. This study aims to determine if elevated UA affects amino acid levels and whether amino acids mediate this relationship, focusing on the causal links between UA, circulating amino acids, and kidney disease. METHODS This study utilized Uox-KO mice as a hyperuricemia model, assessed renal injury through blood biochemistry and pathology, analyzed serum amino acid changes via targeted amino acidomics, and employed Mendelian randomization to investigate the causal links between uric acid, amino acids, and renal disease. RESULTS Hyperuricemia Uox-KO mice have significantly higher serum UA and renal impairment markers, with histopathological analysis showing extensive renal tissue damage. Changes in amino acid balance were found in the mice's serum, with key metabolites like alanine, isoleucine, leucine, aspartic acid, cysteine, glutamate, and glycine potentially influencing UA pathophysiology. Genetically predicted UA was positively correlated with chronic renal failure (CRF) and blood urea nitrogen(BUN) levels and negatively with serum cystatin C (eGFRcys) and serum creatinine (eGFRcrea). Alanine (Ala) mediated the effect of UA on elevated CRF and BUN risk, accounting for 4.5% of the UA-CRF relationship and 14.4% of the UA-BUN association. CONCLUSION In hyperuricemia mice, serum amino acids undergo metabolic changes. Genetically predicted UA levels are positively linked to CRF and BUN, but negatively linked to eGFRcys and eGFRcrea. Ala mediates UA's effect on CRF and BUN risk, indicating Ala could be a target for preventing renal diseases caused by hyperuricemia.
Collapse
Affiliation(s)
- Qinglin Sheng
- University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Yuqing Ma
- Department of Naval Medicine, Naval Medical University, Shanghai, 200093, China
| | - Bingjie Geng
- Department of Naval Medicine, Naval Medical University, Shanghai, 200093, China
| | - Jiahui Chen
- Department of Naval Medicine, Naval Medical University, Shanghai, 200093, China
| | - Junfei Cheng
- Department of Naval Medicine, Naval Medical University, Shanghai, 200093, China
| | - Su Liu
- Department of Naval Medicine, Naval Medical University, Shanghai, 200093, China
| | - Rui Li
- Department of Naval Medicine, Naval Medical University, Shanghai, 200093, China
| | - Xiangtong Li
- Department of Naval Medicine, Naval Medical University, Shanghai, 200093, China
| | - Jing Wang
- Department of Naval Medicine, Naval Medical University, Shanghai, 200093, China
| | - Hongtao Lu
- Department of Naval Medicine, Naval Medical University, Shanghai, 200093, China
| | - Fangyuan Gao
- Department of Naval Medicine, Naval Medical University, Shanghai, 200093, China
| | - Fu Gao
- University of Shanghai for Science and Technology, Shanghai, 200093, China.
- Department of Naval Medicine, Naval Medical University, Shanghai, 200093, China.
| |
Collapse
|
39
|
Zhou T, Lin W, Yang B, Liu Y, Huang W, Xie N, Yang F, Lin Z, Hu Z, Luo S, Luo J. Remnant cholesterol and risk of aortic aneurysm and dissection: a prospective cohort Study from the UK biobank study and mendelian randomization analysis. Lipids Health Dis 2025; 24:53. [PMID: 39962497 PMCID: PMC11831829 DOI: 10.1186/s12944-025-02466-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/04/2025] [Indexed: 02/21/2025] Open
Abstract
AIM This study aimed to examine the relationships between remnant cholesterol (RC) and the risk of aortic aneurysm and dissection (AAD). METHODS This prospective cohort study included 368,139 European adults from the UK Biobank. Additionally, the causal relationship between RC and AAD was investigated using Mendelian randomization (MR) analyses. RESULTS During a median follow-up of 13.65 years, 1,634 cases of abdominal aortic aneurysm (AAA), 698 cases of thoracic aortic aneurysm (TAA), and 184 cases of aortic dissection (AD) were identified. Elevated RC levels were associated with an increased risk of AAA compared to the reference group ([highest vs. lowest RC levels]: adjusted hazard ratio (HR) = 1.65, 95% CI: 1.36-1.99). However, no significant association was observed between high RC levels and the risk of either TAA or AD. Two-sample MR analyses supported a significant causal effect of RC on AAA risk (odds ratio (OR) = 2.08, 95% CI: 1.70-2.56). The association between RC and AAA persisted after adjusting for the effects of RC-associated genetic variants on low-density lipoprotein cholesterol (LDL-C). In contrast, MR analyses did not indicate any causal associations between RC and TAA or AD. CONCLUSIONS Elevated RC was linked to a greater risk of developing AAA, with MR analyses confirming a causal relationship. These findings suggest that RC may function as a new biomarker for AAA and could be integral to strategies aimed at preventing AAA.
Collapse
Affiliation(s)
- Ting Zhou
- Department of Cardiovascular, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Wenhui Lin
- Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial Department of Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Bangyuan Yang
- Department of Cardiovascular, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Yuan Liu
- Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial Department of Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Wenhui Huang
- Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial Department of Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Nianjin Xie
- Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial Department of Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Fan Yang
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zhuoheng Lin
- Department of Cardiovascular, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Ziyang Hu
- Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial Department of Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Songyuan Luo
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Ganzhou Municipal Hospital, Southern Medical University, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Guangzhou, China.
| | - Jianfang Luo
- Guangdong Cardiovascular Institute, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Provincial Department of Cardiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
40
|
Kalwick M, Roth M. A Comprehensive Review of the Genetics of Dyslipidemias and Risk of Atherosclerotic Cardiovascular Disease. Nutrients 2025; 17:659. [PMID: 40004987 PMCID: PMC11858766 DOI: 10.3390/nu17040659] [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: 01/14/2025] [Revised: 02/03/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Dyslipidemias are often diagnosed based on an individual's lipid panel that may or may not include Lp(a) or apoB. But these values alone omit key information that can underestimate risk and misdiagnose disease, which leads to imprecise medical therapies that reduce efficacy with unnecessary adverse events. For example, knowing whether an individual's dyslipidemia is monogenic can granularly inform risk and create opportunities for precision therapeutics. This review explores the canonical and non-canonical causes of dyslipidemias and how they impact atherosclerotic cardiovascular disease (ASCVD) risk. This review emphasizes the multitude of genetic causes that cause primary hypercholesterolemia, hypertriglyceridemia, and low or elevated high-density lipoprotein (HDL)-cholesterol levels. Within each of these sections, this review will explore the evidence linking these genetic conditions with ASCVD risk. Where applicable, this review will summarize approved therapies for a particular genetic condition.
Collapse
Affiliation(s)
| | - Mendel Roth
- GBinsight, GB Healthwatch, San Diego, CA 92122, USA;
| |
Collapse
|
41
|
Fu L, Liu Q, Cheng H, Zhao X, Xiong J, Mi J. Insights Into Causal Effects of Genetically Proxied Lipids and Lipid-Modifying Drug Targets on Cardiometabolic Diseases. J Am Heart Assoc 2025; 14:e038857. [PMID: 39868518 PMCID: PMC12074789 DOI: 10.1161/jaha.124.038857] [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: 09/13/2024] [Accepted: 12/13/2024] [Indexed: 01/28/2025]
Abstract
BACKGROUND The differential impact of serum lipids and their targets for lipid modification on cardiometabolic disease risk is debated. This study used Mendelian randomization to investigate the causal relationships and underlying mechanisms. METHODS Genetic variants related to lipid profiles and targets for lipid modification were sourced from the Global Lipids Genetics Consortium. Summary data for 10 cardiometabolic diseases were compiled from both discovery and replication data sets. Expression quantitative trait loci data from relevant tissues were employed to evaluate significant lipid-modifying drug targets. Comprehensive analyses including colocalization, mediation, and bioinformatics were conducted to validate the results and investigate potential mediators and mechanisms. RESULTS Significant causal associations were identified between lipids, lipid-modifying drug targets, and various cardiometabolic diseases. Notably, genetic enhancement of LPL (lipoprotein lipase) was linked to reduced risks of myocardial infarction (odds ratio [OR]1, 0.65 [95% CI, 0.57-0.75], P1=2.60×10-9; OR2, 0.59 [95% CI, 0.49-0.72], P2=1.52×10-7), ischemic heart disease (OR1, 0.968 [95% CI, 0.962-0.975], P1=5.50×10-23; OR2, 0.64 [95% CI, 0.55-0.73], P2=1.72×10-10), and coronary heart disease (OR1, 0.980 [95% CI, 0.975-0.985], P1=3.63×10-14; OR2, 0.64 [95% CI, 0.54-0.75], P2=6.62×10-8) across 2 data sets. Moreover, significant Mendelian randomization and strong colocalization associations for the expression of LPL in blood and subcutaneous adipose tissue were linked with myocardial infarction (OR, 0.918 [95% CI, 0.872-0.967], P=1.24×10-3; PP.H4, 0.99) and coronary heart disease (OR, 0.991 [95% CI, 0.983-0.999], P=0.041; PP.H4=0.92). Glucose levels and blood pressure were identified as mediators in the total effect of LPL on cardiometabolic outcomes. CONCLUSIONS The study substantiates the causal role of lipids in specific cardiometabolic diseases, highlighting LPL as a potent drug target. The effects of LPL are suggested to be influenced by changes in glucose and blood pressure, providing insights into its mechanism of action.
Collapse
Affiliation(s)
- Liwan Fu
- Center for Non‐Communicable Disease ManagementBeijing Children’s Hospital, Capital Medical University, National Center for Children’s HealthBeijingChina
| | - Qin Liu
- Department of UltrasoundChildren’s Hospital of the Capital Institute of PediatricsBeijingChina
| | - Hong Cheng
- Department of EpidemiologyCapital Institute of PediatricsBeijingChina
| | - Xiaoyuan Zhao
- Department of EpidemiologyCapital Institute of PediatricsBeijingChina
| | - Jingfan Xiong
- Child and Adolescent Chronic Disease Prevention and Control DepartmentShenzhen Center for Chronic Disease ControlShenzhenChina
| | - Jie Mi
- Center for Non‐Communicable Disease ManagementBeijing Children’s Hospital, Capital Medical University, National Center for Children’s HealthBeijingChina
- Key Laboratory of Major Diseases in Children, Ministry of EducationChina
| |
Collapse
|
42
|
Deng Y, Zeng L, Lai Y, Ji S, Peng B, Lu H, Wang M, Kwan HY, Wang Q, Zhao X. Branched-chain amino acids levels associated with risk of erectile dysfunction: A Mendelian randomization analysis. Exp Gerontol 2025; 200:112677. [PMID: 39778693 DOI: 10.1016/j.exger.2025.112677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 12/30/2024] [Accepted: 01/05/2025] [Indexed: 01/11/2025]
Abstract
BACKGROUND Erectile dysfunction (ED) is a prevalent male sexual dysfunction that remarkably impacts patients' quality of life and is also recognized as a precursor to cardiovascular disease (CVD) events. Branched-chain amino acids (BCAAs) are derived from dietary intake and mainly involved in energy metabolism. Previous studies have underscored the association between BCAAs and CVD, but the causal link between BCAAs and ED remains uncertain. METHODS The bidirectional Mendelian randomization (MR) study used the genetic data from genome-wide association studies (GWAS) to identify single nucleotide polymorphisms (SNPs) associated with total BCAAs, leucine, isoleucine, and valine. The genetic data for ED were acquired from the FinnGen study (n = 95,178). The primary method used to assess causal associations was the inverse variance-weighted (IVW) method, supplemented by MR-Egger, weighted median, and simple median analyses. Cochrane's Q test was utilized to evaluate heterogeneity within the results, while the MR-Egger intercept test was utilized to evaluate the Level pleiotropy. A sensitivity analysis was performed employing leave-one-out analysis. RESULTS The MR analysis results indicate a positive correlation between levels of total BCAA (OR = 1.984, 95 % CI = 1.018-3.868, P = 0.044), leucine (OR = 2.277, 95 % CI = 1.121-4.626, P = 0.023), isoleucine (OR = 2.584, 95 % CI = 1.167-5.722, P = 0.019), valine (OR = 1.894, 95 % CI = 1.119-3.206, P = 0.017), and the risk of ED. Sensitivity tests confirmed the accuracy and robustness of the study findings. Moreover, the reverse MR analysis found no association between ED and the BCAAs. CONCLUSION The results of this analysis indicate a positive association between the circulating BCAA concentrations and the risk of ED, but their underlying mechanisms require further investigation.
Collapse
Affiliation(s)
- Yijian Deng
- Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Liying Zeng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yigui Lai
- People's Hospital of Yangjiang, Yangjiang 529500, China
| | - Shuai Ji
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Baizhao Peng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Hanqi Lu
- Dongguan Hospital of Traditional Chinese Medicine, Dongguan, Guangdong 523000, China
| | - Ming Wang
- Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Hiu Yee Kwan
- School of Chinese Medicine, Hong Kong Baptist University, Hong Kong, China
| | - Qi Wang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China.
| | - Xiaoshan Zhao
- Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
43
|
Yang LZ, Yang Y, Hong C, Wu QZ, Shi XJ, Liu YL, Chen GZ. Systematic Mendelian Randomization Exploring Druggable Genes for Hemorrhagic Strokes. Mol Neurobiol 2025; 62:1359-1372. [PMID: 38977622 PMCID: PMC11772512 DOI: 10.1007/s12035-024-04336-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 06/26/2024] [Indexed: 07/10/2024]
Abstract
Patients with hemorrhagic stroke have high rates of morbidity and mortality, and drugs for prevention are very limited. Mendelian randomization (MR) analysis can increase the success rate of drug development by providing genetic evidence. Previous MR analyses only analyzed the role of individual drug target genes in hemorrhagic stroke; therefore, we used MR analysis to systematically explore the druggable genes for hemorrhagic stroke. We sequentially performed summary-data-based MR analysis and two-sample MR analysis to assess the associations of all genes within the database with intracranial aneurysm, intracerebral hemorrhage, and their subtypes. Validated genes were further analyzed by colocalization. Only genes that were positive in all three analyses and were druggable were considered desirable genes. We also explored the mediators of genes affecting hemorrhagic stroke incidence. Finally, the associations of druggable genes with other cardiovascular diseases were analyzed to assess potential side effects. We identified 56 genes that significantly affected hemorrhagic stroke incidence. Moreover, TNFSF12, SLC22A4, SPARC, KL, RELT, and ADORA3 were found to be druggable. The inhibition of TNFSF12, SLC22A4, and SPARC can reduce the risk of intracranial aneurysm, subarachnoid hemorrhage, and intracerebral hemorrhage. Gene-induced hypertension may be a potential mechanism by which these genes cause hemorrhagic stroke. We also found that blocking these genes may cause side effects, such as ischemic stroke and its subtypes. Our study revealed that six druggable genes were associated with hemorrhagic stroke, and the inhibition of TNFSF12, SLC22A4, and SPARC had preventive effects against hemorrhagic strokes.
Collapse
Affiliation(s)
- Lun-Zhe Yang
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yong Yang
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Chuan Hong
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qi-Zhe Wu
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xiong-Jie Shi
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yi-Lin Liu
- Department of Neurosurgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Guang-Zhong Chen
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
44
|
Lyu H, Fan N, Wen H, Zhang X, Mao H, Bian Q, Chen J. Interplay between BMI, neutrophil, triglyceride and uric acid: a case-control study and bidirectional multivariate mendelian randomization analysis. Nutr Metab (Lond) 2025; 22:7. [PMID: 39876024 PMCID: PMC11776270 DOI: 10.1186/s12986-025-00896-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 01/08/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND This study aims to explore the interplay between body mass index (BMI), neutrophils, triglyceride levels, and uric acid (UA). Understanding the causal correlation between UA and health indicators, specifically its association with the body's inflammatory conditions, is crucial for preventing and managing various diseases. METHODS A retrospective analysis was conducted on 4,286 cases utilizing the Spearman correlation method. BMI, neutrophil count, and triglyceride levels were determined as key exposure factors. To further investigate the causal correlation, a two-sample Mendelian randomization(MR) design was utilized, leveraging data from genome-wide association study (GWAS). Within this framework, and multivariable Mendelian randomization(MVMR) was applied to explore the linkage between multiple genetic variants and complex traits. RESULTS The study primarily focused on UA, employing genetic variation as a natural tool to assess the causal impact of various factors on UA. Spearman correlation analysis revealed significant association between UA and BMI (ρ = 0.230,p<0.01), neutrophils (ρ = 0.164,p<0.01), and triglyceride levels (ρ = 0.154,p<0.01). Additionally, two-sample MR analysis affirmed a reciprocal causal association between neutrophils and UA (OR = 1.035,95%CI:1.009-1.061,p = 0.008), as well as positive causal connection between UA and both BMI (OR = 1.083,95%CI:1.042-1.126,p<0.001) and triglyceride levels (OR = 1.090,95%CI:1.037-1.146,p<0.001). Neutrophils also demonstrated a positive causal linkage with BMI (OR = 1.034,95%CI:1.009-1.078,p = 0.012) and triglyceride levels (OR = 1.077,95%CI:1.033-1.122,p<0.001), and BMI exhibited a similar causal association with triglyceride levels (OR = 1.300,95%CI:1.212-1.385,p<0.001). These findings shed light on the causal networks connecting UA, BMI, neutrophils, and triglyceride levels. By integrating Spearman correlation analysis with various MR study designs, this study provided a robust framework for identifying key factors influencing hyperuricemia and related health issues, thereby enhancing our understanding of the interplay between inflammatory markers and these health indicators. CONCLUSIONS Our study presents strong evidence of the complex interconnection between BMI, neutrophils, triglyceride, and UA, revealing complex causal links and highlighting potential inflammatory states as key mediators. The findings may contribute to a better understanding of these factors and potentially lead to improved clinical outcomes and patients' health.
Collapse
Affiliation(s)
- Haoyuan Lyu
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China
- Huangjiahu Hospital, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China
- Hubei Shizhen Laboratory, Wuhan, Hubei, 430065, China
| | - Na Fan
- Hubei Shizhen Laboratory, Wuhan, Hubei, 430065, China
- School of Physical Education and Health Sciences, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China
| | - Hao Wen
- First Clinical College, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China
- Hubei Shizhen Laboratory, Wuhan, Hubei, 430065, China
| | - Xin Zhang
- Hubei Shizhen Laboratory, Wuhan, Hubei, 430065, China
- School of Physical Education and Health Sciences, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China
| | - Herong Mao
- School of Foreign Languages, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China
- Hubei Shizhen Laboratory, Wuhan, Hubei, 430065, China
| | - Qinglai Bian
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China
- Hubei Shizhen Laboratory, Wuhan, Hubei, 430065, China
| | - Jiaxu Chen
- School of Basic Medical Sciences, Hubei University of Chinese Medicine, Wuhan, Hubei, 430065, China.
- Guangzhou Key Laboratory of Formula-Pattern of Traditional Chinese Medicine, Jinan University, Guangzhou, Guangdong, 510632, China.
- Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, Hubei, 430065, China.
- Hubei Shizhen Laboratory, Wuhan, Hubei, 430065, China.
| |
Collapse
|
45
|
Khan SR, Ye WW, Van JAD, Singh I, Rabiee Y, Rodricks KL, Zhang X, Nicholson RJ, Razani B, Summers SA, Futerman AH, Gunderson EP, Wheeler MB. Reduced circulating sphingolipids and CERS2 activity are linked to T2D risk and impaired insulin secretion. SCIENCE ADVANCES 2025; 11:eadr1725. [PMID: 39792658 PMCID: PMC11790001 DOI: 10.1126/sciadv.adr1725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 12/09/2024] [Indexed: 01/12/2025]
Abstract
Gestational diabetes mellitus (GDM), a transient form of diabetes that resolves postpartum, is a major risk factor for type 2 diabetes (T2D) in women. While the progression from GDM to T2D is not fully understood, it involves both genetic and environmental components. By integrating clinical, metabolomic, and genome-wide association study (GWAS) data, we identified associations between decreased sphingolipid biosynthesis and future T2D, in part through the rs267738 allele of the CERS2 gene in Hispanic women shortly after a GDM pregnancy. To understand the impact of the CERS2 gene and risk allele on glucose regulation, we examined whole-body Cers2 knockout and rs267738 knock-in mice. Both models exhibited glucose intolerance and impaired insulin secretion in vivo. Islets isolated from these models also demonstrated reduced β cell function, as shown by decreased insulin secretion ex vivo. Overall, reduced circulating sphingolipids may indicate a high risk of GDM-to-T2D progression and reflect deficits in CERS2 activity that negatively affect glucose homeostasis and β cell function.
Collapse
Affiliation(s)
- Saifur R. Khan
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- VA Medical Center, Pittsburgh, PA, USA
- Center for Immunometabolism, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wenyue W. Ye
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Julie A. D. Van
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Ishnoor Singh
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Yasmin Rabiee
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | | | - Xiangyu Zhang
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- VA Medical Center, Pittsburgh, PA, USA
- Center for Immunometabolism, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rebekah J. Nicholson
- Departments of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Babak Razani
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA, USA
- VA Medical Center, Pittsburgh, PA, USA
- Center for Immunometabolism, University of Pittsburgh, Pittsburgh, PA, USA
| | - Scott A. Summers
- Departments of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, USA
| | - Anthony H. Futerman
- Department of Biomolecular Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Erica P. Gunderson
- Division of Research, Kaiser Permanente Northern California, Pleasanton, CA, USA
- Department of Health Systems Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA, USA
| | - Michael B. Wheeler
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
46
|
Zhao F, Yang Y, Yang W. Exploring the causal impact of body mass index on metabolic biomarkers and cholelithiasis risk: a Mendelian randomization analysis. Sci Rep 2025; 15:415. [PMID: 39747165 PMCID: PMC11697197 DOI: 10.1038/s41598-024-83217-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025] Open
Abstract
Obesity is a well-established risk factor for various diseases, but the mechanisms through which it influences disease development remain unclear. Using Mendelian randomization (MR) analysis, we examined the causal relationship between BMI, 249 metabolic traits, and cholelithiasis. BMI data were obtained from four sources, and cholelithiasis data were from two distinct datasets. We analyzed the direct effect of BMI on cholelithiasis and identified key metabolic mediators. BMI was found to be positively associated with the risk of cholelithiasis across all datasets analyzed. A total of 176 metabolites were identified to be significantly associated with BMI, including amino acids, cholesterol esters, free cholesterol, triglycerides, and phospholipids. Among these, 49 metabolites were identified as mediators in the BMI-cholelithiasis relationship. Specifically, fatty acid levels, cholesteryl esters, phospholipids, triglycerides, and free cholesterol were key mediators in this relationship, with mediation proportions ranging from - 2.38-7.14%. This study provides robust evidence that BMI significantly impacts metabolic biomarkers, which in turn affect the risk of cholelithiasis. These findings highlight the importance of managing BMI to mitigate metabolic dysfunction and reduce the risk of gallstone formation. Future research should explore the specific metabolic pathways involved to identify potential therapeutic targets.
Collapse
Affiliation(s)
- Feng Zhao
- The First Hospital of Anhui University of Science & Technology (Huainan First People's Hospital), Huainan, 232000, Anhui Province, China
| | - Yanjiang Yang
- Department of Rheumatology and Immunology, The people's Hospital of Qiandongnan Autonomous Prefecture, Kaili, 556000, Guizhou Province, China
| | - Wenwen Yang
- The First Clinical Medical College, Lanzhou University, Lanzhou, 730000, Gansu Province, China.
| |
Collapse
|
47
|
Yang Y, Wang X, Yang W. Exploring the influence of cheese consumption on blood metabolites: Implications for disease pathogenesis, with a focus on essential hypertension. J Dairy Sci 2025; 108:119-135. [PMID: 39477058 DOI: 10.3168/jds.2024-25585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/05/2024] [Indexed: 12/28/2024]
Abstract
This study examines the causal effects of cheese consumption on blood metabolites and essential hypertension using Mendelian randomization (MR). Genome-wide association study data from the Open GWAS project and FinnGen Biobank were analyzed, focusing on cheese intake, 249 metabolic traits, and hypertension. Instrumental variables were selected based on strict criteria, and causal relationships were assessed using inverse-variance weighing, weighted-median, and MR-Egger methods. Cheese consumption significantly influenced 118 metabolic traits, including AA, cholesterol esters, and triglycerides. Notably, it was linked to a reduction in hypertension risk (odds ratio = 0.652; 95% CI: 0.541-0.785). Mediation analysis identified 50 metabolic traits as intermediaries, accounting for 0.88% to 8.25% of the total effect. These findings suggest that moderate cheese intake may benefit cardiovascular health by lowering hypertension risk, emphasizing the importance of cheese type and dietary context in health recommendations.
Collapse
Affiliation(s)
- Yanjiang Yang
- Department of Rheumatology and Immunology, The People's Hospital of Qiandongnan Autonomous Prefecture, Kaili 556000, Guizhou Province, China
| | - Xiaorui Wang
- School of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Wenwen Yang
- The First Clinical Medical College, Lanzhou University, Lanzhou 730000, Gansu Province, China.
| |
Collapse
|
48
|
Modder M, het Panhuis WI, Li M, Afkir S, Dorn AL, Pronk ACM, Streefland TCM, Lalai RA, Pierrou S, Nilsson SK, Olivecrona G, Kooijman S, Rensen PCN, Schönke M. Liver-targeted Angptl4 silencing by antisense oligonucleotide treatment attenuates hyperlipidaemia and atherosclerosis development in APOE*3-Leiden.CETP mice. Cardiovasc Res 2024; 120:2179-2190. [PMID: 39259836 PMCID: PMC11687395 DOI: 10.1093/cvr/cvae195] [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: 06/06/2024] [Accepted: 06/19/2024] [Indexed: 09/13/2024] Open
Abstract
AIMS Angiopoietin-like 3 (ANGPTL3) and 4 (ANGPTL4) inhibit lipoprotein lipase to regulate tissue fatty acid (FA) uptake from triglyceride (TG)-rich lipoproteins such as very low density lipoproteins (VLDL). While pharmacological inhibition of ANGPTL3 is being evaluated as a lipid-lowering strategy, systemic ANGPTL4 inhibition is not pursued due to adverse effects. This study aims to compare the therapeutic potential of liver-specific Angptl3 and Angptl4 silencing to attenuate hyperlipidemia and atherosclerosis development in APOE*3-Leiden.CETP mice, a well-established humanized model for lipoprotein metabolism. METHODS AND RESULTS Mice were subcutaneously injected twice per week with saline or liver-targeted antisense oligonucleotides against Angptl3, Angptl4, both, or a scrambled oligonucleotide. Plasma lipid levels, VLDL clearance, and hepatic VLDL production were determined, and atherosclerosis development was assessed. For toxicological evaluation, cynomolgus monkeys were treated with three dosages of liver-targeted ANGPTL4-silencing oligonucleotides. Liver-targeted Angptl4 silencing reduced plasma TGs (-48%) and total cholesterol (-56%), explained by higher VLDL-derived FA uptake by brown adipose tissue and lower VLDL production by the liver. Accordingly, Angptl4 silencing reduced atherosclerotic lesion size (-86%) and improved lesion stability. Hepatic Angptl3 silencing similarly attenuated hyperlipidemia and atherosclerosis development. While Angptl3 and Angptl4 silencing lowered plasma TGs in the refed and fasted state, respectively, combined Angptl3/4 silencing lowered plasma TGs independent of the nutritional state. In cynomolgus monkeys, anti-ANGPTL4 ASO treatment was well tolerated without adverse effects. CONCLUSION Liver-targeted Angptl4 silencing potently attenuates hyperlipidemia and atherosclerosis development in APOE*3-Leiden.CETP mice, and liver-targeted ANGPTL4 silencing is well tolerated in non-human primates. These data warrant further clinical development of liver-targeted ANGPTL4 silencing.
Collapse
Affiliation(s)
- Melanie Modder
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Wietse In het Panhuis
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Mohan Li
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Salwa Afkir
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Alexandra L Dorn
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Amanda C M Pronk
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Trea C M Streefland
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Reshma A Lalai
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Stefan Pierrou
- Lipigon Pharmaceuticals AB, Tvistevägen 48C, 907 36 Umeå, Sweden
| | - Stefan K Nilsson
- Lipigon Pharmaceuticals AB, Tvistevägen 48C, 907 36 Umeå, Sweden
| | - Gunilla Olivecrona
- Lipigon Pharmaceuticals AB, Tvistevägen 48C, 907 36 Umeå, Sweden
- Department of Medical Biosciences, Umeå University, Umeå, Sweden
| | - Sander Kooijman
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Milena Schönke
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
49
|
Huang H, Qin J, Wen Z, Wang C, Chen C, Liu Y, Li H, Cao S, Yang X. Association of branched-chain amino acids and risk of three urologic cancers: a Mendelian randomization study. Transl Cancer Res 2024; 13:6709-6720. [PMID: 39816560 PMCID: PMC11729756 DOI: 10.21037/tcr-24-1142] [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: 07/04/2024] [Accepted: 10/31/2024] [Indexed: 01/18/2025]
Abstract
Background Multiple studies suggest a plausible connection between urologic cancers and branched-chain amino acids (BCAAs) breakdown metabolic enzymes. Nevertheless, there is scarce exploration into the variations in circulating BCAAs. In our research, we utilize bidirectional, two-sample Mendelian randomization (MR) analysis to predict the link between BCAAs levels and three distinct types of urological tumors. Methods The study examined data from the UK Biobank, including a comprehensive genome-wide association study (GWAS) of total BCAAs, leucine, isoleucine, and valine, alongside three urological system tumors [prostate cancer (PCa), kidney cancer, and bladder cancer] sourced from the Medical Research Council Integrative Epidemiology Unit (MRC-IEU) and FinnGen Consortium databases. The primary analytical approach involved the use of the inverse variance weighted (IVW) method, complemented by MR-PRESSO global testing and MR-Egger regression to identify potential horizontal pleiotropy. Heterogeneity was evaluated using the Cochran Q test. Results The levels of circulating total BCAAs [odds ratio (OR) =1.002688, 95% confidence interval (CI): 1.000, 1.005, P=0.03], leucine (OR =1.0038, 95% CI: 1.001, 1.007, P=0.008), isoleucine (OR =1.003352, 95% CI: 1.000, 1.007, P=0.04), and valine (OR =1.00279, 95% CI: 1.001, 1.005, P=0.009) showed positive associations with PCa risk. However, there was inadequate evidence to establish a link between BCAAs and bladder or kidney cancer. Conclusions In summary, an association existed between elevated levels of circulating total BCAAs, leucine, isoleucine, and valine, and an increased risk of PCa. However, no correlation was detected between BCAAs and kidney or bladder cancer.
Collapse
Affiliation(s)
- Haotian Huang
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jiao Qin
- Department of Anesthesiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Zhi Wen
- Department of Urology, Langzhong People’s Hospital, Langzhong, China
| | - Chongjian Wang
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Caixia Chen
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yang Liu
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Hongyuan Li
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Song Cao
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xuesong Yang
- Department of Urology, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
- Health Management Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
50
|
Liu S, Fu Z, Liu H, Wang Y, Zhou M, Ding Z, Feng Z. Lipid Profiles, Telomere Length, and the Risk of Malignant Tumors: A Mendelian Randomization and Mediation Analysis. Biomedicines 2024; 13:13. [PMID: 39857597 PMCID: PMC11760878 DOI: 10.3390/biomedicines13010013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/15/2024] [Accepted: 12/19/2024] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: The relationship between lipid profiles, telomere length (TL), and cancer risk remains unclear. Methods: This study employed two-sample Mendelian randomization (MR) with mediation analysis to investigate their causal relationships, examining lipid profiles as exposure, TL as mediator, and nine cancer types as outcomes. We conducted our analysis using two-stage least squares (2SLS) regression integrated with inverse variance weighted (IVW) methods to address potential endogeneity and strengthen our causal inference. Results: we found that unfavorable lipid profiles were causally linked to increased TL (p < 0.05). TL showed positive causal associations with lung and hematologic cancers (OR > 1, p < 0.05). Direct associations were observed between total and low-density lipoprotein (LDL) cholesterol and gastric cancer (OR < 1, p < 0.05), and between remnant cholesterol and colorectal cancer (OR > 1, p < 0.05). Mediation analysis revealed TL as a significant mediator in the pathway from lipid profiles to cancer development (p < 0.05). No horizontal pleiotropy was detected. Conclusions: Our findings suggest that lipid metabolism disorders may influence cancer development through telomere regulation, particularly in lung and hematologic cancers. This emphasizes the importance of lipid management in cancer prevention and treatment, especially for these cancer types.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhenhua Ding
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Radiation Medicine, School of Public Health, Southern Medical University, Guangzhou 510515, China; (S.L.); (Z.F.); (H.L.); (Y.W.); (M.Z.)
| | - Zhijun Feng
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Radiation Medicine, School of Public Health, Southern Medical University, Guangzhou 510515, China; (S.L.); (Z.F.); (H.L.); (Y.W.); (M.Z.)
| |
Collapse
|