201
|
Atak M, Sevim Nalkiran H, Bostan M, Uydu HA. The association of Sort1 expression with LDL subfraction and inflammation in patients with coronary artery disease. Acta Cardiol 2024; 79:159-166. [PMID: 38095557 DOI: 10.1080/00015385.2023.2285534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 11/14/2023] [Indexed: 04/18/2024]
Abstract
BACKGROUND Controversial effect of sortilin on lipoprotein metabolism in the development of atherosclerosis reveals the need for more extensive research. OBJECTIVES The aim of this study was to investigate the association between Sort1 gene expression and lipids, lipoprotein subfractions, and inflammation in CAD. METHODS The study population included 162 subjects with CAD and 49 healthy individuals. The Sort1 gene expression level was determined by qRT-PCR using Human Sortilin TaqMan Gene Expression Assays. Lipoprotein subclasses were analysed by the Lipoprint system. Serum levels of apolipoprotein and CRP were measured by autoanalyzer. RESULTS Sort1 gene expression and atherogenic subfraction (SdLDL) levels were significantly higher (p < 0.001) while atheroprotective subfraction (LbLDL) was lower in the subjects with CAD (p < 0.050). Also, increased Sort1 gene expression levels were observed in those with higher CRP values. CONCLUSIONS Our findings reveal that the high Sort1 gene expression has a prominent linear relationship with both the atherogenic LDL phenotype and proinflammation, thereby might contribute to the occurrence of CAD.
Collapse
Affiliation(s)
- Mehtap Atak
- Recep Tayyip Erdogan University, Rize, Turkey
| | | | | | | |
Collapse
|
202
|
Yan Z, Xu Y, Li K, Liu L. Association between genetically proxied lipid-lowering drug targets, lipid traits, and amyotrophic lateral sclerosis: a mendelian randomization study. Acta Neurol Belg 2024; 124:485-494. [PMID: 37889424 DOI: 10.1007/s13760-023-02393-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 09/21/2023] [Indexed: 10/28/2023]
Abstract
BACKGROUND The use of circulating lipid traits as biomarkers to predict the risk of amyotrophic lateral sclerosis (ALS) is currently controversial, and the evidence-based medical evidence for the use of lipid-lowering agents, especially statins, on ALS risk remains insufficient. Our aim was to apply a Mendelian randomization (MR) approach to assess the causal impact of lipid-lowering agents and circulating lipid traits on ALS risk. MATERIALS AND METHODS Our study included primary and secondary analyses, in which the risk associations of lipid-lowering gene inhibitors, lipid traits, and ALS were assessed by the inverse variance weighting method as the primary approach. The robustness of the results was assessed using LDSC assessment, conventional MR sensitivity analysis, and used Mediating MR to explore potential mechanisms of occurrence. In the secondary analysis, the association of lipid-lowering genes with ALS was validated using the Summary data-based Mendelian Randomization (SMR) method. RESULTS Our results showed strong evidence between genetic proxies for Apolipoprotein B (ApoB) inhibitor (OR = 0.76, 95% CI = 0.68 - 0.86; P = 5.58 × 10-6) and reduced risk of ALS. Additionally, 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) inhibitor (OR = 1.06, 95% CI = 0. 85-1.33) was not found to increase ALS risk. SMR results suggested that ApoB expression was associated with increased ALS risk, and colocalization analysis did not support a significant common genetic variation between ApoB and ALS. Mediator MR analysis suggested a possible mediating role for interleukin-6 and low-density lipoprotein cholesterol (LDL-C). While elevated LDL-C was significantly associated with increased risk of ALS among lipid traits, total cholesterol (TC) and ApoB were weakly associated with ALS. LDSC results suggested a potential genetic correlation between these lipid traits and ALS. CONCLUSIONS Using ApoB inhibitor can lower the risk of ALS, statins do not trigger ALS, and LDL-C, TC, and ApoB levels can predict the risk of ALS.
Collapse
Affiliation(s)
- Zhaoqi Yan
- Graduate School, Jiangxi University of Traditional Chinese Medicine, Yangming Road, Nanchang, Jiangxi, China
| | - Yifeng Xu
- Graduate School, Jiangxi University of Traditional Chinese Medicine, Yangming Road, Nanchang, Jiangxi, China
| | - Keke Li
- Graduate School, Jiangxi University of Traditional Chinese Medicine, Yangming Road, Nanchang, Jiangxi, China
| | - Liangji Liu
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, 445 Bayi Dadao, Nanchang, Jiangxi, China.
| |
Collapse
|
203
|
Zou J, Qi S, Sun X, Zhang Y, Wang Y, Li Y, Zhao ZH, Lei D. Association of lipid-modifying therapy with risk of obstructive sleep apnea: A drug-target mendelian randomization study. Toxicol Appl Pharmacol 2024; 485:116909. [PMID: 38521370 DOI: 10.1016/j.taap.2024.116909] [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: 02/12/2024] [Revised: 03/11/2024] [Accepted: 03/19/2024] [Indexed: 03/25/2024]
Abstract
BACKGROUND Obstructive sleep apnea (OSA) is considered to be an important contributor of dyslipidemia. However, there lacks observational studies focusing on the potential effect of lipid management on OSA risk. Thus, we aimed to investigate the genetic association of lipid-modifying therapy with risk of OSA. METHODS A drug-target mendelian randomization (MR) study using both cis-variants and cis-expression quantitative trait loci (eQTLs) of lipid-modifying drug targets was performed. The MR analyses used summary-level data of genome wide association studies (GWAS). Primary MR analysis was conducted using inverse-variance-weighted (IVW) method. Sensitivity analysis was performed using weighted median (WM) and MR-pleiotropy residual sum and outlier (MR-PRESSO) methods. RESULTS Genetically proxied low-density lipoprotein cholesterol (LDL-C)-lowering effect of cholesteryl ester transfer protein (CETP) was associated with reduced risk of OSA (odds ratio [OR] =0.75, 95% confidence interval [CI]: 0.60-0.94, false discovery rate [FDR] q value = 0.046). A significant MR association with risk of OSA was observed for CETP expression in subcutaneous adipose tissue (OR = 0.94, 95%CI: 0.89-1.00, FDR q value = 0.049), lung (OR = 0.94, 95%CI: 0.89-1.00, FDR q value = 0.049) and small intestine (OR = 0.96, 95%CI: 0.93-1.00, FDR q value = 0.049). No significant effects of high-density lipoprotein cholesterol (HDL-C)-raising effect of CETP inhibition, LDL-C-lowering and triglycerides-lowering effect of other drug targets on OSA risk were observed. CONCLUSIONS The present study presented genetic evidence supporting the association of LDL-C-lowering therapy by CETP inhibition with reduced risk of OSA. These findings provided novel insights into the role of lipid management in patients with OSA and encouraged further clinical validations and mechanistic investigations.
Collapse
Affiliation(s)
- Juanjuan Zou
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, NHC Key Laboratory of Otorhinolaryngology (Shandong University), Jinan 250012, China; Medical Integration and Practice Center, Shandong University, Jinan 250012, China
| | - Shengnan Qi
- Department of Pathology, Qingdao Eighth People's Hospital, Qingdao 266000, China
| | - Xiaojing Sun
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, NHC Key Laboratory of Otorhinolaryngology (Shandong University), Jinan 250012, China
| | - Yijing Zhang
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, NHC Key Laboratory of Otorhinolaryngology (Shandong University), Jinan 250012, China
| | - Yan Wang
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, NHC Key Laboratory of Otorhinolaryngology (Shandong University), Jinan 250012, China
| | - Yanzhong Li
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, NHC Key Laboratory of Otorhinolaryngology (Shandong University), Jinan 250012, China
| | - Ze-Hua Zhao
- Department of Hepatology, Qilu Hospital of Shandong University, Jinan 250012, China.
| | - Dapeng Lei
- Department of Otorhinolaryngology, Qilu Hospital of Shandong University, NHC Key Laboratory of Otorhinolaryngology (Shandong University), Jinan 250012, China.
| |
Collapse
|
204
|
Pang X, Yang H, Li M, Suarez-Farinas M, Tian S. To explore the causal association between the serum lipid profile and inflammatory bowel disease using bidirectional Mendelian randomisation analysis. EGASTROENTEROLOGY 2024; 2:e100034. [PMID: 39944468 PMCID: PMC11770443 DOI: 10.1136/egastro-2023-100034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 05/03/2024] [Indexed: 04/01/2025]
Abstract
Background Despite studies confirming that patients with inflammatory bowel disease (IBD) present with dyslipidaemia, the associations between IBD and the serum lipid profile have not been determined. The present study aimed to investigate the causal relationship between the serum lipid profile and IBD risk and elucidate the nature of the interactions between them. Methods Two-sample Mendelian randomisation (MR) analysis was performed to investigate the causal links between total cholesterol (TC), total triglyceride (TG), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), apolipoprotein A (Apo A), apolipoprotein B (Apo B) and lipoprotein (a) (Lp(a)) and IBD. The study was carried out using the R TwoSampleMR and Mendelian randomisation packages. Results All MR methods, including the weighted median, weighted mode, inverse-variance weighted model, MR-PRESSO, contamination mixture and MR Egger, supported a null causal relationship between TG, TC, HDL-C, LDL-C, Apo A, Apo B and Lp(a) and between IBD, Crohn's disease and ulcerative colitis. Null causal effects of lipid indices on IBD were validated through independent genome-wide association studies (GWAS), indicating that the findings are robust. Conclusion Our findings suggest that none of the seven lipid indices may be a potential risk factor for the onset of IBD. However, additional research is needed since our MR analyses cannot assess the potential non-linear causal relationship between serum lipids and IBD.
Collapse
Affiliation(s)
- Xiaoli Pang
- Department of Pediatric Gastroenterology, Children's Medical Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Huizhong Yang
- Department of Pediatric Gastroenterology, Children's Medical Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Mingyu Li
- Department of Family Planning, Jilin Women and Children Health Hospital, Changchun, Jilin, China
| | - Mayte Suarez-Farinas
- Department of Population Health Science & Policy, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, New York City, New York, USA
| | - Suyan Tian
- Division of Clinical Research, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
205
|
Ji X, Guo HY, Han M, Peng H, Yuan H. Association between genetically proxied PCSK9 inhibition and systemic lupus erythematosus risk: A mendelian randomization study. Int J Rheum Dis 2024; 27:e15106. [PMID: 38568054 DOI: 10.1111/1756-185x.15106] [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: 10/18/2023] [Revised: 12/13/2023] [Accepted: 02/21/2024] [Indexed: 04/05/2024]
Abstract
BACKGROUND Preclinical and epidemiological studies suggest that proprotein convertase subtilisin/kexin type 9 (PCSK9) had a potential effect on the development of SLE, but it was unclear whether a causal relationship exists. We aimed to investigate the association between genetically proxied inhibition of PCSK9 and the risk of SLE using a two-sample Mendelian randomization (MR) approach. METHODS Single nucleotide polymorphisms (SNPs) associated with PCSK9 were extracted from pooled data obtained from the Global Lipid Genetics Consortium (GLGC) Genome-wide Association Study (GWAS) related to LDL-c levels, which was used as a proxy for PCSK9 inhibition. Pooled statistics for SLE were obtained from an independent GWAS dataset including 5201 SLE patients and 9066 controls. Inverse variance-weighted random-effects models were used to examine the association between genetically proxied inhibition of PCSK9 and the risk of SLE. MR-Egger, weighted median, weighted mode, Simple mode, and co-location analyses were used as sensitivity analyses to test the robustness of the analyses. RESULTS Genetically proxied inhibition of PCSK9 was associated with a reduced risk of SLE (OR = 0.51, 95% CI = 0.34 to 0.77, p = .001). This finding was replicated in an earlier GLGC GWAS analysis (OR = 0.59, 95% CI = 0.40 to 0.87, p = .007). Sensitivity analysis ensured that the results were robust. Co-localization analysis did not find evidence of shared causal variation between PCSK9 and SLE. CONCLUSIONS This Mendelian randomization study showed that PCSK9 was associated with SLE pathogenesis, and its inhibition was associated with a reduced risk of SLE. This study has offered a prospective therapeutic avenue for intervening in the progression of SLE by inhibiting PCSK9 levels.
Collapse
Affiliation(s)
- Xincan Ji
- School of Public Health, Wannan Medical College, Wuhu, Anhui, China
| | - Hao-Yang Guo
- School of Public Health, Wannan Medical College, Wuhu, Anhui, China
| | - Mengqi Han
- School of Public Health, Wannan Medical College, Wuhu, Anhui, China
| | - Hui Peng
- Science and Technology Department, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, China
| | - Hui Yuan
- School of Public Health, Wannan Medical College, Wuhu, Anhui, China
| |
Collapse
|
206
|
Hu X, Chen F, Jia L, Long A, Peng Y, Li X, Huang J, Wei X, Fang X, Gao Z, Zhang M, Liu X, Chen YG, Wang Y, Zhang H, Wang Y. A gut-derived hormone regulates cholesterol metabolism. Cell 2024; 187:1685-1700.e18. [PMID: 38503280 DOI: 10.1016/j.cell.2024.02.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 11/18/2023] [Accepted: 02/21/2024] [Indexed: 03/21/2024]
Abstract
The reciprocal coordination between cholesterol absorption in the intestine and de novo cholesterol synthesis in the liver is essential for maintaining cholesterol homeostasis, yet the mechanisms governing the opposing regulation of these processes remain poorly understood. Here, we identify a hormone, Cholesin, which is capable of inhibiting cholesterol synthesis in the liver, leading to a reduction in circulating cholesterol levels. Cholesin is encoded by a gene with a previously unknown function (C7orf50 in humans; 3110082I17Rik in mice). It is secreted from the intestine in response to cholesterol absorption and binds to GPR146, an orphan G-protein-coupled receptor, exerting antagonistic downstream effects by inhibiting PKA signaling and thereby suppressing SREBP2-controlled cholesterol synthesis in the liver. Therefore, our results demonstrate that the Cholesin-GPR146 axis mediates the inhibitory effect of intestinal cholesterol absorption on hepatic cholesterol synthesis. This discovered hormone, Cholesin, holds promise as an effective agent in combating hypercholesterolemia and atherosclerosis.
Collapse
Affiliation(s)
- Xiaoli Hu
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Fengyi Chen
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Liangjie Jia
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Aijun Long
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Ying Peng
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xu Li
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Junfeng Huang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xueyun Wei
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Xinlei Fang
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zihua Gao
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Mengxian Zhang
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Xiao Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Ye-Guang Chen
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China; Guangzhou Laboratory, Guangzhou 510005, China; School of Basic Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Yan Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Huijie Zhang
- Department of Endocrinology and Metabolism, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China.
| | - Yiguo Wang
- State Key Laboratory of Membrane Biology, MOE Key Laboratory of Bioinformatics, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
207
|
Li F, Mei Y, Wu Q, Wu X. Drug Target Mendelian Randomization Study of PCSK9 and HMG-CoA Reductase Inhibition and Atrial Fibrillation. Cardiology 2024; 149:495-501. [PMID: 38531334 PMCID: PMC11449189 DOI: 10.1159/000538551] [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: 10/15/2023] [Accepted: 03/23/2024] [Indexed: 03/28/2024]
Abstract
INTRODUCTION Atrial fibrillation (AF) is a prevalent cardiac arrhythmia with significant clinical implications. The potential influence of lipid-lowering therapies, specifically PCSK9 inhibitors (PCSK9i) and HMG-CoA reductase inhibitors (statins), on AF risk remains a topic of interest. This mendelian randomization (MR) study aimed to elucidate the causal relationship between genetically predicted inhibition of PCSK9 and HMG-CoA reductase and the risk of AF. METHODS Utilizing publicly available, summary-level genome-wide association study data, we employed single-nucleotide polymorphisms associated with lower LDL-C levels as instruments for gene-simulated inhibition of PCSK9 and HMG-CoA reductase. Multiple MR techniques were applied to estimate the causal effects, and sensitivity analyses were conducted to validate the results. RESULTS Genetically predicted inhibition of PCSK9 demonstrated a reduced risk of AF, with an odds ratio (OR) of 0.92 (95% CI: 0.85-0.99, p = 0.01) using the inverse variance-weighted (IVW) method. In contrast, the inhibition of HMG-CoA reductase did not exhibit a statistically significant association with AF risk (IVW: OR = 1.11, 95% CI: 1.00-1.22, p = 0.05). CONCLUSION Our MR study suggests that genetically predicted inhibition of PCSK9, but not HMG-CoA reductase, is associated with a lower risk of AF. These findings provide evidence for a causal protective effect of PCSK9i on AF and support the use of PCSK9i for AF prevention in patients with dyslipidemia. Further studies are needed to elucidate the mechanisms underlying the differential effects of PCSK9i and statins on AF and to confirm the clinical implications of our findings.
Collapse
Affiliation(s)
- Fuyuan Li
- Department of Cardiology, Lishui People's Hospital, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, China
- Department of Cardiology, First Affiliated Hospital of Lishui University School of Medicine, Lishui, China
| | - Yibin Mei
- Department of Cardiology, Lishui People's Hospital, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, China
- Department of Cardiology, First Affiliated Hospital of Lishui University School of Medicine, Lishui, China
| | - Qiongbi Wu
- Department of Cardiology, Lishui People's Hospital, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, China
- Department of Cardiology, First Affiliated Hospital of Lishui University School of Medicine, Lishui, China
| | - Xianjun Wu
- Department of Cardiology, Lishui People's Hospital, The Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, China
- Department of Cardiology, First Affiliated Hospital of Lishui University School of Medicine, Lishui, China
| |
Collapse
|
208
|
Li Z, Wang W, Li W, Duan H, Xu C, Tian X, Ning F, Zhang D. Co-methylation analyses identify CpGs associated with lipid traits in Chinese discordant monozygotic twins. Hum Mol Genet 2024; 33:583-593. [PMID: 38142287 DOI: 10.1093/hmg/ddad207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/29/2023] [Accepted: 12/05/2023] [Indexed: 12/25/2023] Open
Abstract
To control genetic background and early life milieu in genome-wide DNA methylation analysis for blood lipids, we recruited Chinese discordant monozygotic twins to explore the relationships between DNA methylations and total cholesterol (TC), high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol (LDL-C), and triglycerides (TG). 132 monozygotic (MZ) twins were included with discordant lipid levels and completed data. A linear mixed model was conducted in Epigenome-wide association study (EWAS). Generalized estimating equation model was for gene expression analysis. We conducted Weighted correlation network analysis (WGCNA) to build co-methylated interconnected network. Additional Qingdao citizens were recruited for validation. Inference about Causation through Examination of Familial Confounding (ICE FALCON) was used to infer the possible direction of these relationships. A total of 476 top CpGs reached suggestively significant level (P < 10-4), of which, 192 CpGs were significantly associated with TG (FDR < 0.05). They were used to build interconnected network and highlight crucial genes from WGCNA. Finally, four CpGs in GATA4 were validated as risk factors for TC; six CpGs at ITFG2-AS1 were negatively associated with TG; two CpGs in PLXND1 played protective roles in HDL-C. ICE FALCON indicated abnormal TC was regarded as the consequence of DNA methylation in CpGs at GATA4, rather than vice versa. Four CpGs in ITFG2-AS1 were both causes and consequences of modified TG levels. Our results indicated that DNA methylation levels of 12 CpGs in GATA4, ITFG2-AS1, and PLXND1 were relevant to TC, TG, and HDL-C, respectively, which might provide new epigenetic insights into potential clinical treatment of dyslipidemia.
Collapse
Affiliation(s)
- Zhaoying Li
- Department of Epidemiology and Health Statistics, The College of Public Health of Qingdao University, No. 308 Ning Xia Street, Qingdao 266071, Shandong Province, People's Republic of China
| | - Weijing Wang
- Department of Epidemiology and Health Statistics, The College of Public Health of Qingdao University, No. 308 Ning Xia Street, Qingdao 266071, Shandong Province, People's Republic of China
| | - Weilong Li
- Epidemiology and Biostatistics, Department of Public Health, University of Southern Denmark, J.B. Winsløws Vej 9 B, st. tv. Odense C DK-5000, Denmark
| | - Haiping Duan
- Qingdao Municipal Center for Disease Control and Prevention, No. 175 Shandong Road, Qingdao 266000, Shandong Province, People's Republic of China
- Qingdao Institute of Preventive Medicine, No. 175 Shandong Road, Qingdao 266000, Shandong Province, People's Republic of China
| | - Chunsheng Xu
- Qingdao Municipal Center for Disease Control and Prevention, No. 175 Shandong Road, Qingdao 266000, Shandong Province, People's Republic of China
- Qingdao Institute of Preventive Medicine, No. 175 Shandong Road, Qingdao 266000, Shandong Province, People's Republic of China
| | - Xiaocao Tian
- Qingdao Municipal Center for Disease Control and Prevention, No. 175 Shandong Road, Qingdao 266000, Shandong Province, People's Republic of China
- Qingdao Institute of Preventive Medicine, No. 175 Shandong Road, Qingdao 266000, Shandong Province, People's Republic of China
| | - Feng Ning
- Qingdao Municipal Center for Disease Control and Prevention, No. 175 Shandong Road, Qingdao 266000, Shandong Province, People's Republic of China
- Qingdao Institute of Preventive Medicine, No. 175 Shandong Road, Qingdao 266000, Shandong Province, People's Republic of China
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, The College of Public Health of Qingdao University, No. 308 Ning Xia Street, Qingdao 266071, Shandong Province, People's Republic of China
| |
Collapse
|
209
|
Wang Z, Wang X, Shi Y, Wu S, Ding Y, Yao G, Chen J. Advancements in elucidating the pathogenesis of actinic keratosis: present state and future prospects. Front Med (Lausanne) 2024; 11:1330491. [PMID: 38566927 PMCID: PMC10985158 DOI: 10.3389/fmed.2024.1330491] [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: 10/30/2023] [Accepted: 02/19/2024] [Indexed: 04/04/2024] Open
Abstract
Solar keratosis, also known as actinic keratosis (AK), is becoming increasingly prevalent. It is a benign tumor that develops in the epidermis. Individuals with AK typically exhibit irregular, red, scaly bumps or patches as a result of prolonged exposure to UV rays. These growths primarily appear on sun-exposed areas of the skin such as the face, scalp, and hands. Presently, dermatologists are actively studying AK due to its rising incidence rate in the United States. However, the underlying causes of AK remain poorly understood. Previous research has indicated that the onset of AK involves various mechanisms including UV ray-induced inflammation, oxidative stress, complex mutagenesis, resulting immunosuppression, inhibited apoptosis, dysregulated cell cycle, altered cell proliferation, tissue remodeling, and human papillomavirus (HPV) infection. AK can develop in three ways: spontaneous regression, persistence, or progression into invasive cutaneous squamous cell carcinoma (cSCC). Multiple risk factors and diverse signaling pathways collectively contribute to its complex pathogenesis. To mitigate the risk of cancerous changes associated with long-term UV radiation exposure, prompt identification, management, and prevention of AK are crucial. The objective of this review is to elucidate the primary mechanisms underlying AK malignancy and identify potential treatment targets for dermatologists in clinical settings.
Collapse
Affiliation(s)
- Zhongzhi Wang
- Department of Dermatology, Shanghai Fourth People’s Hospital, Tongji University, Shanghai, China
| | - Xiaolie Wang
- Department of Dermatology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Yuanyang Shi
- Department of Dermatology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Siyu Wu
- Department of Dermatology, Shanghai Fourth People’s Hospital, Tongji University, Shanghai, China
| | - Yu Ding
- Department of Dermatology, Shanghai Fourth People’s Hospital, Tongji University, Shanghai, China
| | - Guotai Yao
- Department of Dermatology, Changzheng Hospital, Naval Medical University, Shanghai, China
| | - Jianghan Chen
- Department of Dermatology, Shanghai Fourth People’s Hospital, Tongji University, Shanghai, China
- Department of Dermatology, Naval Medical Center, Naval Medical University, Shanghai, China
| |
Collapse
|
210
|
Zhang Y, Ren E, Zhang C, Wang Y, Chen X, Li L. The protective role of oily fish intake against type 2 diabetes: insights from a genetic correlation and Mendelian randomization study. Front Nutr 2024; 11:1288886. [PMID: 38567249 PMCID: PMC10986736 DOI: 10.3389/fnut.2024.1288886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 03/11/2024] [Indexed: 04/04/2024] Open
Abstract
Background and aims Previous research has underscored the association between oily fish intake and type 2 diabetes (T2DM), yet the causality remains elusive. Methods A bidirectional univariable Mendelian Randomization (MR) analysis was employed to evaluate the causal effects of oily fish and non-oily fish intake on T2DM. Replication analysis and meta-analysis were conducted to ensure robust results. Multivariable MR analysis was utilized to assess confounders, and further mediation MR analysis discerned mediating effects. Linkage Disequilibrium Score (LDSC) analysis was undertaken to compute genetic correlations. Inverse variance weighted (IVW) was the primary method, complemented by a series of sensitivity analyses. Results The LDSC analysis unveiled a significant genetic correlation between oily fish intake and T2DM (Genetic correlation: -0.102, p = 4.43 × 10-4). For each standard deviation (SD) increase in genetically predicted oily fish intake, the risk of T2DM was reduced by 38.6% (OR = 0.614, 95% CI 0.504 ~ 0.748, p = 1.24 × 10-6, False Discovery Rate (FDR) = 3.72 × 10-6). The meta-analysis across three data sources highlighted a persistent causal association (OR = 0.728, 95% CI 0.593 ~ 0.895, p = 0.003). No other causal effects were identified (all p > 0.5, FDR > 0.5). The main outcomes remained consistent in most sensitivity analyses. Both MVMR and mediation MR analyses emphasized the mediating roles of triglycerides (TG), body mass index (BMI), and 25-hydroxyvitamin D (25OHD) levels. Conclusion To encapsulate, there's an inverse association between oily fish intake and T2DM risk, suggesting potential benefits of oily fish intake in T2DM prevention.
Collapse
Affiliation(s)
- Youqian Zhang
- Department of Endocrinology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
- Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Entong Ren
- Health Science Center, Yangtze University, Jingzhou, Hubei, China
- Southern Theater General Hospital, Guangzhou, Guangdong, China
| | - Chunlong Zhang
- Health Science Center, Yangtze University, Jingzhou, Hubei, China
- Department of Nursing, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Yang Wang
- Department of Neurology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaohe Chen
- Health Science Center, Yangtze University, Jingzhou, Hubei, China
| | - Lin Li
- Department of Endocrinology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
211
|
Zhang F, Yu Z. Mendelian randomization study on insulin resistance and risk of hypertension and cardiovascular disease. Sci Rep 2024; 14:6191. [PMID: 38485964 PMCID: PMC10940700 DOI: 10.1038/s41598-023-46983-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 01/07/2022] [Indexed: 03/18/2024] Open
Abstract
Observational studies have suggested that insulin resistance (IR) is associated with hypertension and various cardiovascular diseases. However, the presence of a causal relationship between IR and cardiovascular disease remains unclear. Here, we applied Mendelian randomization (MR) approaches to address the causal association between genetically determined IR and the risk of cardiovascular diseases. Our primary genetic instruments comprised 53 SNPs associated with IR phenotype from a GWAS of up to 188,577 participants. Genetic association estimates for hypertension and venous thromboembolism (VTE) were extracted from UK Biobank, estimates for atrial fibrillation (AF) were extracted from the hitherto largest GWAS meta-analysis on AF, estimates for heart failure were extracted from HERMES Consortium, estimates for peripheral artery disease (PAD) and aortic aneurysm were extracted from the FinnGen Study. The main analyses were performed using the random-effects inverse-variance weighted approach, and complemented by sensitivity analyses and multivariable MR analyses. Corresponding to 55% higher fasting insulin adjusted for body mass index, 0.46 mmol/L lower high-density lipoprotein cholesterol and 0.89 mmol/L higher triglyceride, one standard deviation change in genetically predicted IR was associated with increased risk of hypertension (odds ratio (OR) 1.06, 95% CI 1.04-1.08; P = 1.91 × 10-11) and PAD (OR 1.90, 95% CI 1.43-2.54; P = 1.19 × 10-5). Suggestive evidence was obtained for an association between IR and heart failure (OR per SD change in IR: 1.19, 95% CI 1.01-1.41, P = 0.041). There was no MR evidence for an association between genetically predicted IR and atrial fibrillation, VTE, and aortic aneurysm. Results were widely consistent across all sensitivity analyses. In multivariable MR, the association between IR and PAD was attenuated after adjustment for lipids (P = 0.347) or BMI (P = 0.163). Our findings support that genetically determined IR increases the risk of hypertension and PAD.
Collapse
Affiliation(s)
- Fangfang Zhang
- Department of Outpatient, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Zhimin Yu
- Department of Geriatrics, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China.
| |
Collapse
|
212
|
Huang S, Liu Y, Zhang Y, Wang Y, Gao Y, Li R, Yu L, Hu X, Fang Q. Analyzing the causal relationship between lipid-lowering drug target genes and epilepsy: a Mendelian randomization study. Front Neurol 2024; 15:1331537. [PMID: 38523609 PMCID: PMC10957583 DOI: 10.3389/fneur.2024.1331537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 02/15/2024] [Indexed: 03/26/2024] Open
Abstract
Background Previous research has yielded conflicting results on the link between epilepsy risk and lipid-lowering medications. The aim of this study is to determine whether the risk of epilepsy outcomes is causally related to lipid-lowering medications predicted by genetics. Methods We used genetic instruments as proxies to the exposure of lipid-lowering drugs, employing variants within or near genes targeted by these drugs and associated with low-density lipoprotein cholesterol (LDL cholesterol) from a genome-wide association study. These variants served as controlling factors. Through drug target Mendelian randomization, we systematically assessed the impact of lipid-lowering medications, including HMG-CoA reductase (HMGCR) inhibitors, proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, and Niemann-Pick C1-like 1 (NPC1L1) inhibitors, on epilepsy. Results The analysis demonstrated that a higher expression of HMGCR was associated with an elevated risk of various types of epilepsy, including all types (OR = 1.17, 95% CI:1.03 to 1.32, p = 0.01), focal epilepsy (OR = 1.24, 95% CI:1.08 to 1.43, p = 0.003), and focal epilepsy documented with lesions other than hippocampal sclerosis (OR = 1.05, 95% CI: 1.01 to 1.10, p = 0.02). The risk of juvenile absence epilepsy (JAE) was also associated with higher expression of PCSK9 (OR = 1.06, 95% CI: 1.02 to 1.09, p = 0.002). For other relationships, there was no reliable supporting data available. Conclusion The drug target MR investigation suggests a possible link between reduced epilepsy vulnerability and HMGCR and PCSK9 inhibition.
Collapse
Affiliation(s)
- Shicun Huang
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuan Liu
- Department of Neurology, Suzhou Ninth People’s Hospital, Suzhou, China
| | - Yi Zhang
- Department of Neurology, The Affiliated Changzhou NO.2 People’s Hospital of Nanjing Medical University, Changzhou, China
| | - Yiqing Wang
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ya Gao
- Department of Neurology, Suzhou Guangci Cancer Hospital, Suzhou, China
| | - Runnan Li
- Department of Neurology, The Dushu Lake Hospital of Soochow University, Suzhou, China
| | - Lidong Yu
- Department of Neurology, The Affiliated Taizhou Second People’s Hospital of Yangzhou University, Yangzhou, China
| | - Xiaowei Hu
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, China
| | - Qi Fang
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
213
|
Jiang Y, Zhang W, Wei M, Yin D, Tang Y, Jia W, Wang C, Guo J, Li A, Gong Y. Associations between type 1 diabetes and pulmonary tuberculosis: a bidirectional mendelian randomization study. Diabetol Metab Syndr 2024; 16:60. [PMID: 38443967 PMCID: PMC10913601 DOI: 10.1186/s13098-024-01296-x] [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: 11/23/2023] [Accepted: 02/20/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Type 1 diabetes mellitus (T1DM) has been associated with higher pulmonary tuberculosis (PTB) risk in observational studies. However, the causal relationship between them remains unclear. This study aimed to assess the causal effect between T1DM and PTB using bidirectional Mendelian randomization (MR) analysis. METHODS Single nucleotide polymorphisms (SNPs) of T1DM and PTB were extracted from the public genetic variation summary database. In addition, GWAS data were collected to explore the causal relationship between PTB and relevant clinical traits of T1DM, including glycemic traits, lipids, and obesity. The inverse variance weighting method (IVW), weighted median method, and MR‒Egger regression were used to evaluate the causal relationship. To ensure the stability of the results, sensitivity analyses assess the robustness of the results by estimating heterogeneity and pleiotropy. RESULTS IVW showed that T1DM increased the risk of PTB (OR = 1.07, 95% CI: 1.03-1.12, P < 0.001), which was similar to the results of MR‒Egger and weighted median analyses. Moreover, we found that high-density lipoprotein cholesterol (HDL-C; OR = 1.28, 95% CI: 1.03-1.59, P = 0.026) was associated with PTB. There was no evidence of an effect of glycemic traits, remaining lipid markers, or obesity on the risk of PTB. In the reverse MR analysis, no causal relationships were detected for PTB on T1DM and its relevant clinical traits. CONCLUSION This study supported that T1DM and HDL-C were risk factors for PTB. This implies the effective role of treating T1DM and managing HDL-C in reducing the risk of PTB, which provides an essential basis for the prevention and comanagement of concurrent T1DM and PTB in clinical practice.
Collapse
Affiliation(s)
- Yijia Jiang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, 100700, Beijing, China
| | - Wenhua Zhang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, 100700, Beijing, China
| | - Maoying Wei
- Dongzhimen Hospital, Beijing University of Chinese Medicine, 100700, Beijing, China
| | - Dan Yin
- Dongzhimen Hospital, Beijing University of Chinese Medicine, 100700, Beijing, China
| | - Yiting Tang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, 100700, Beijing, China
| | - Weiyu Jia
- Dongzhimen Hospital, Beijing University of Chinese Medicine, 100700, Beijing, China
| | - Churan Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, 100700, Beijing, China
| | - Jingyi Guo
- Dongzhimen Hospital, Beijing University of Chinese Medicine, 100700, Beijing, China
| | - Aijing Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, 100700, Beijing, China
| | - Yanbing Gong
- Dongzhimen Hospital, Beijing University of Chinese Medicine, 100700, Beijing, China.
| |
Collapse
|
214
|
Woerner J, Sriram V, Nam Y, Verma A, Kim D. Uncovering genetic associations in the human diseasome using an endophenotype-augmented disease network. Bioinformatics 2024; 40:btae126. [PMID: 38527901 PMCID: PMC10963079 DOI: 10.1093/bioinformatics/btae126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 01/17/2024] [Indexed: 03/27/2024] Open
Abstract
MOTIVATION Many diseases, particularly cardiometabolic disorders, exhibit complex multimorbidities with one another. An intuitive way to model the connections between phenotypes is with a disease-disease network (DDN), where nodes represent diseases and edges represent associations, such as shared single-nucleotide polymorphisms (SNPs), between pairs of diseases. To gain further genetic understanding of molecular contributors to disease associations, we propose a novel version of the shared-SNP DDN (ssDDN), denoted as ssDDN+, which includes connections between diseases derived from genetic correlations with intermediate endophenotypes. We hypothesize that a ssDDN+ can provide complementary information to the disease connections in a ssDDN, yielding insight into the role of clinical laboratory measurements in disease interactions. RESULTS Using PheWAS summary statistics from the UK Biobank, we constructed a ssDDN+ revealing hundreds of genetic correlations between diseases and quantitative traits. Our augmented network uncovers genetic associations across different disease categories, connects relevant cardiometabolic diseases, and highlights specific biomarkers that are associated with cross-phenotype associations. Out of the 31 clinical measurements under consideration, HDL-C connects the greatest number of diseases and is strongly associated with both type 2 diabetes and heart failure. Triglycerides, another blood lipid with known genetic causes in non-mendelian diseases, also adds a substantial number of edges to the ssDDN. This work demonstrates how association with clinical biomarkers can better explain the shared genetics between cardiometabolic disorders. Our study can facilitate future network-based investigations of cross-phenotype associations involving pleiotropy and genetic heterogeneity, potentially uncovering sources of missing heritability in multimorbidities. AVAILABILITY AND IMPLEMENTATION The generated ssDDN+ can be explored at https://hdpm.biomedinfolab.com/ddn/biomarkerDDN.
Collapse
Affiliation(s)
- Jakob Woerner
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Vivek Sriram
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Genomics and Computational Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Yonghyun Nam
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Anurag Verma
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA 19104, United States
| | - Dokyoon Kim
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, United States
- Institute for Biomedical Informatics, University of Pennsylvania, Philadelphia, PA 19104, United States
| |
Collapse
|
215
|
Liang Y, Deng MG, Jian Q, Liu M, Fang K, Chen S. Maternal history of Alzheimer's disease predisposes to altered serum cholesterol levels in adult offspring. J Neurochem 2024; 168:303-311. [PMID: 38316937 DOI: 10.1111/jnc.16056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 01/08/2024] [Accepted: 01/08/2024] [Indexed: 02/07/2024]
Abstract
Controversial findings regarding the association between serum cholesterol levels and Alzheimer's disease (AD) have been identified through observational studies. The genetic basis shared by both factors and the causality between them remain largely unknown. The objective of this study is to examine the causal impact of maternal history of AD on changes in serum cholesterol levels in adult offspring. By retrieving genetic variants from summary statistics of large-scale genome-wide association study of maternal history of AD (European-based: Ncase = 27 696, Ncontrol = 260 980). The causal association between genetically predicted maternal history of AD and changes in serum cholesterol levels in adult offspring was examined using the two-sample Mendelian randomization (MR) method. Causal impact estimates were calculated using single-nucleotide polymorphisms in both univariable MR (UMR) and multivariable MR (MVMR) analyses. Additionally, other approaches, such as Cochran's Q test and leave-one-out variant analysis, were employed to correct for potential biases. The results of UMR presented that genetically predicted maternal history of AD was positively associated with hypercholesterolemia (OR = 1.014; 95% CI: 1.009-1.018; p < 0.001), total cholesterol (OR = 1.29; 95% CI: 1.134-1.466; p < 0.001) and low-density lipoprotein (OR = 1.525; 95% CI: 1.272-1.828; p < 0.001) among adult offspring. Genetic predisposition for maternal history of AD to be negatively associated with high-density lipoprotein (OR = 0.889; 95% CI: 0.861-0.917; p < 0.001). The MVMR analysis remained robust and significant after adjusting for diabetes and obesity in offspring. Sufficient evidence was provided in this study to support the putative causal impact of maternal history of AD on the change of serum cholesterol profile in adult offspring. In clinical practice, priority should be given to the detection and monitoring of cholesterol levels in individuals with a maternal history of AD, particularly in the early stages.
Collapse
Affiliation(s)
- Yuehui Liang
- School of Public Health, Wuhan University, Wuhan, China
| | - Ming-Gang Deng
- Department of Psychiatry, Wuhan Mental Health Centre, Wuhan, China
- Department of Psychiatry, Wuhan Hospital for Psychotherapy, Wuhan, China
| | - Qinghong Jian
- The Affiliated Stomatology Hospital of Southwest Medical University, Luzhou, China
| | - Mingwei Liu
- School of Public Health, Wuhan University, Wuhan, China
- Julius Global Health, The Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Kui Fang
- Department of Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Shuai Chen
- School of Public Health, Wuhan University, Wuhan, China
| |
Collapse
|
216
|
Riesmeijer SA, Nolte IM, Olde Loohuis LM, Reus LM, Boltz T, Ng M, Furniss D, Werker PMN, Ophoff RA. Polygenic Risk Associations with Clinical Characteristics and Recurrence of Dupuytren Disease. Plast Reconstr Surg 2024; 153:573e-583e. [PMID: 37257093 PMCID: PMC10876167 DOI: 10.1097/prs.0000000000010775] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/22/2023] [Indexed: 06/02/2023]
Abstract
BACKGROUND Dupuytren disease (DD) is a common complex trait, with varying severity and incompletely understood cause. Genome-wide association studies (GWAS) have identified risk loci. In this article, we examine whether genetic risk profiles of DD in patients are associated with clinical variation and disease severity and with patient genetic risk profiles of genetically correlated traits, including body mass index (BMI), triglycerides, high-density lipoproteins, type 2 diabetes mellitus, and endophenotypes fasting glucose and glycated hemoglobin. METHODS The authors used a well-characterized cohort of 1461 DD patients with available phenotypic and genetic data. Phenotype data include age at onset, recurrence, and family history of disease. Polygenic risk scores (PRSs) of DD, BMI, triglycerides, high-density lipoprotein, type 2 diabetes, fasting glucose, and hemoglobin A1c using various significance thresholds were calculated with PRSice using the most recent GWAS summary statistics. Control data from LifeLines were used to determine P value cutoffs for PRS generation explaining most variance. RESULTS The PRS for DD was significantly associated with a positive family history for DD, age at onset, disease onset before the age of 50, and recurrence. We also found a significant negative correlation between the PRSs for DD and BMI. CONCLUSIONS Although GWAS studies of DD are designed to identify genetic risk factors distinguishing case/control status, we show that the genetic risk profile for DD also explains part of its clinical variation and disease severity. The PRS may therefore aid in accurate prognostication, choosing initial treatment and in personalized medicine in the future. CLINICAL QUESTION/LEVEL OF EVIDENCE Risk, III.
Collapse
Affiliation(s)
- Sophie A. Riesmeijer
- From the Departments of Plastic Surgery
- Epidemiology, University of Groningen, University Medical Center Groningen
| | - Ilja M. Nolte
- Epidemiology, University of Groningen, University Medical Center Groningen
| | - Loes M. Olde Loohuis
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles
| | - Lianne M. Reus
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam University Medical Center
| | - Toni Boltz
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles
| | - Michael Ng
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford
| | - Dominic Furniss
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford
| | | | - Roel A. Ophoff
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles
- Department of Psychiatry, Erasmus University Rotterdam, Erasmus Medical Center
| |
Collapse
|
217
|
Votava JA, John SV, Li Z, Chen S, Fan J, Parks BW. Mining cholesterol genes from thousands of mouse livers identifies aldolase C as a regulator of cholesterol biosynthesis. J Lipid Res 2024; 65:100525. [PMID: 38417553 PMCID: PMC10965479 DOI: 10.1016/j.jlr.2024.100525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 02/12/2024] [Accepted: 02/16/2024] [Indexed: 03/01/2024] Open
Abstract
The availability of genome-wide transcriptomic and proteomic datasets is ever-increasing and often not used beyond initial publication. Here, we applied module-based coexpression network analysis to a comprehensive catalog of 35 mouse genome-wide liver expression datasets (encompassing more than 3800 mice) with the goal of identifying and validating unknown genes involved in cholesterol metabolism. From these 35 datasets, we identified a conserved module of genes enriched with cholesterol biosynthetic genes. Using a systematic approach across the 35 datasets, we identified three genes (Rdh11, Echdc1, and Aldoc) with no known role in cholesterol metabolism. We then performed functional validation studies and show that each gene is capable of regulating cholesterol metabolism. For the glycolytic gene, Aldoc, we demonstrate that it contributes to de novo cholesterol biosynthesis and regulates cholesterol and triglyceride levels in mice. As Aldoc is located within a genome-wide significant genome-wide association studies locus for human plasma cholesterol levels, our studies establish Aldoc as a causal gene within this locus. Through our work, we develop a framework for leveraging mouse genome-wide liver datasets for identifying and validating genes involved in cholesterol metabolism.
Collapse
Affiliation(s)
- James A Votava
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Zhonggang Li
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Shuyang Chen
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Jing Fan
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA; Morgridge Institute for Research, Madison, WI, USA
| | - Brian W Parks
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
218
|
Tang AS, Rankin KP, Cerono G, Miramontes S, Mills H, Roger J, Zeng B, Nelson C, Soman K, Woldemariam S, Li Y, Lee A, Bove R, Glymour M, Aghaeepour N, Oskotsky TT, Miller Z, Allen IE, Sanders SJ, Baranzini S, Sirota M. Leveraging electronic health records and knowledge networks for Alzheimer's disease prediction and sex-specific biological insights. NATURE AGING 2024; 4:379-395. [PMID: 38383858 PMCID: PMC10950787 DOI: 10.1038/s43587-024-00573-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 01/19/2024] [Indexed: 02/23/2024]
Abstract
Identification of Alzheimer's disease (AD) onset risk can facilitate interventions before irreversible disease progression. We demonstrate that electronic health records from the University of California, San Francisco, followed by knowledge networks (for example, SPOKE) allow for (1) prediction of AD onset and (2) prioritization of biological hypotheses, and (3) contextualization of sex dimorphism. We trained random forest models and predicted AD onset on a cohort of 749 individuals with AD and 250,545 controls with a mean area under the receiver operating characteristic of 0.72 (7 years prior) to 0.81 (1 day prior). We further harnessed matched cohort models to identify conditions with predictive power before AD onset. Knowledge networks highlight shared genes between multiple top predictors and AD (for example, APOE, ACTB, IL6 and INS). Genetic colocalization analysis supports AD association with hyperlipidemia at the APOE locus, as well as a stronger female AD association with osteoporosis at a locus near MS4A6A. We therefore show how clinical data can be utilized for early AD prediction and identification of personalized biological hypotheses.
Collapse
Affiliation(s)
- Alice S Tang
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA.
- Graduate Program in Bioengineering, University of California, San Francisco and University of California, Berkeley, San Francisco and Berkeley, CA, USA.
| | - Katherine P Rankin
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Gabriel Cerono
- Weill Institute for Neuroscience. Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Silvia Miramontes
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Hunter Mills
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Jacquelyn Roger
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Billy Zeng
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Charlotte Nelson
- Weill Institute for Neuroscience. Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Karthik Soman
- Weill Institute for Neuroscience. Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Sarah Woldemariam
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Yaqiao Li
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Albert Lee
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Riley Bove
- Weill Institute for Neuroscience. Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Maria Glymour
- Department of Anesthesiology, Pain, and Perioperative Medicine, Stanford University, Palo Alto, CA, USA
| | - Nima Aghaeepour
- Department of Anesthesiology, Pain, and Perioperative Medicine, Stanford University, Palo Alto, CA, USA
- Department of Pediatrics, Stanford University, Palo Alto, CA, USA
- Department of Biomedical Data Science, Stanford University, Palo Alto, CA, USA
| | - Tomiko T Oskotsky
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Zachary Miller
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Isabel E Allen
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, CA, USA
| | - Stephan J Sanders
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
- Institute of Developmental and Regenerative Medicine, Department of Paediatrics, University of Oxford, Oxford, UK
- Department of Psychiatry and Behavioral Sciences, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA
| | - Sergio Baranzini
- Weill Institute for Neuroscience. Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA.
- Department of Pediatrics, University of California, San Francisco, CA, USA.
| |
Collapse
|
219
|
Hu M, Li B, Yang T, Yang Y, Yin C. Effect of Household Income on Cardiovascular Diseases, Cardiovascular Biomarkers, and Socioeconomic Factors. Clin Ther 2024; 46:239-245. [PMID: 38350757 DOI: 10.1016/j.clinthera.2024.01.005] [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: 06/09/2023] [Revised: 09/07/2023] [Accepted: 01/08/2024] [Indexed: 02/15/2024]
Abstract
PURPOSE To examine whether household income is causally related to cardiovascular diseases and investigate the potential reasons. METHODS Using 2-sample Mendelian randomization analyses, we obtained summary statistics from genome-wide association studies of household income and a range of cardiovascular diseases, biomarkers, and socioeconomic factors. FINDINGS Higher household income was causally associated with lower risks of coronary heart disease (odd ratio [OR] = 0.63; 95% CI: 0.49-0.79; P = 0.0001), myocardial infarction (OR = 0.64; 95% CI: 0.50-0.82; P = 0.0003), and hypertension (OR = 0.71; 95% CI: 0.58-0.88; P = 0.0015). With increasing household income, the cardiovascular biomarkers including triglycerides, C-reactive protein, body mass index, fasting glucose were decreased whereas telomere length and high-density lipoprotein cholesterol were increased. Besides, individuals with higher household income were less likely to smoke (β = -0.34; 95% CI: -0.47 to -0.21; P = 1.91×10-07), intake salt (β = -0.14; 95% CI: -0.21 to -0.07; P = 0.0001), or be exposed to air pollution (β = -0.10; 95% CI: -0.15 to -0.06; P = 8.81×10-06) or depression state (β = -0.03; 95% CI: -0.04 to -0.02; P = 5.16×10-07). They were more likely to take physical activity (β = 0.06; 95% CI: 0.02 to 010; P = 0.0016) and have long years of schooling (β = 0.70; 95% CI: 0.62 to 0.78; P = 5.32×10-67). IMPLICATIONS Higher household income is causally associated with better socioeconomic factors and improved cardiovascular biomarkers, which translates into a reduced prevalence of cardiovascular diseases. Policies to improve income equality may result in a reduced burden of cardiovascular diseases.
Collapse
Affiliation(s)
- Mengjin Hu
- Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Boyu Li
- Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tao Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuejin Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Chunlin Yin
- Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
220
|
Qiao L, Lv S, Meng K, Yang J. Genetically proxied therapeutic inhibition of lipid-lowering drug targets and risk of rheumatoid arthritis disease: a Mendelian randomization study. Clin Rheumatol 2024; 43:939-947. [PMID: 38198113 DOI: 10.1007/s10067-023-06837-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 11/12/2023] [Accepted: 11/27/2023] [Indexed: 01/11/2024]
Abstract
OBJECTIVE To evaluate the potential impact of consistent use of similar treatments over a long period; it is essential to investigate the potential correlation between genetic variations that influence the expression or function of pharmacological targets for reducing lipid levels and the risk of developing rheumatoid arthritis. METHODS We used variants in the following genes to conduct Mendelian randomization analyses: HMGCR (encoding the target for statins), PCSK9 (encoding the target for PCSK9 inhibitors, such as evolocumab and alirocumab), and NPC1L1 (encoding the target for ezetimibe). Data from lipid genetics consortia (173,082 sample size) were used to weight variations according to their correlations with low-density lipoprotein cholesterol (LDL-C). In two large datasets (total n = 19,562 cases, 501,655 controls). We conducted a meta-analysis of Mendelian randomization estimates, weighted by LDL-C levels, on the regional differences in the risk of rheumatoid arthritis using data from two large databases. RESULTS We approached SMR and IVW-MR analyses to examine the relationship between target gene expression (including HMGCR, PCSK9, and NPC1L1) and LDL-C levels mediated by these genes with RA. The IVW-MR analysis revealed no significant association between genetically predicted LDL-C concentration and the risk of RA (OR = 0.88, 95% CI = 0.59-1.29; OR = 0.91, 95% CI = 0.67-1.23; OR = 0.81, 95% CI = 0.49-1.36; all p > 0.05). Similarly, our findings from the SMR approach provided no evidence to suggest that gene expression of HMGCR, PCSK9, and NPC1L1 was associated with the risk of RA (OR = 0.91, 95% CI = 0.79-1.05, p = 0.207; OR = 0.96, 95% CI = 0.85-1.09, p = 0.493). CONCLUSIONS Our results do not provide evidence to support the hypothesis that reducing LDL-C levels with statins, alirocumab, or ezetimibe effectively prevents the risk of developing RA. However, our study provides valuable insights into the assessment of lipid-lowering agents in RA, which can enhance our understanding of the condition and assist in clinical practice by aiding in the determination and monitoring of RA status to clinical response.
Collapse
Affiliation(s)
- Liang Qiao
- Shanghai Xuhui District Central Hospital, Shanghai, China
| | - Shun Lv
- Shanghai Xuhui District Central Hospital, Shanghai, China
| | - Kai Meng
- Shanghai Xuhui District Central Hospital, Shanghai, China
| | - Jianmei Yang
- Shanghai Xuhui District Central Hospital, Shanghai, China.
| |
Collapse
|
221
|
Yin X, Wu Y, Song J. Investigating the causal relationship between human blood/urine metabolites and periodontal disease using two-sample Mendelian randomization. Health Sci Rep 2024; 7:e1895. [PMID: 38469110 PMCID: PMC10925816 DOI: 10.1002/hsr2.1895] [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: 08/08/2023] [Revised: 11/07/2023] [Accepted: 01/31/2024] [Indexed: 03/13/2024] Open
Abstract
Background and Aims The aim is to investigate the cause-and-effect connection between metabolites found in blood/urine and the likelihood of developing periodontal disease (PD) through the utilization of a two-sample Mendelian randomization (MR) method. Methods Using an inverse variance weighted (IVW) method and two additional two-sample MR models, we examined the relationship between blood/urine metabolites and PD by analyzing data from a comprehensive metabolome-based genome-wide association study and the Genome-Wide Association Studies (GWAS) of PD. To assess the consistency and dependability of the findings, diversity, cross-effects, and sensitivity analyses were conducted. Results Out of the 35 metabolites found in blood and urine, a total of eight metabolites (C-reactive protein, Potassium in urine, Urea, Cystatin C, Non-albumin protein, Creatinine, estimated Glomerular Filtration Rate, and Phosphate) displayed a possible causal connection with the risk of dental caries/PD using the inverse variance weighted (IVW) method (p < 0.05). This includes five metabolites in the blood and three in the urine. No metabolites were statistically significant in IVW MR models (p < 3.68 × 10- 4). Even after conducting sensitivity analysis with the leave-one-out method and removing the confounding instrumental variables, the impact of these factors on dental caries/PD remained significant. Conclusion Based on the available evidence, it is not possible to establish a significant causal link between the 35 blood metabolites and the likelihood of developing dental caries and PD.
Collapse
Affiliation(s)
- Xinhai Yin
- Department of Oral and Maxillofacial SurgeryGuizhou Provincial People's HospitalGuiyangChina
| | - Yadong Wu
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Guizhou Medical UniversityGuiyangChina
| | - Jukun Song
- Department of Oral and Maxillofacial SurgeryThe Affiliated Stomatological Hospital of Guizhou Medical UniversityGuiyangChina
| |
Collapse
|
222
|
Huang Z, Cui T, Yao J, Wu Y, Zhu J, Yang X, Cui L, Zhou H. Potential association of genetically predicted lipid and lipid-modifying drugs with rheumatoid arthritis: A Mendelian randomization study. PLoS One 2024; 19:e0298629. [PMID: 38416767 PMCID: PMC10901327 DOI: 10.1371/journal.pone.0298629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 01/27/2024] [Indexed: 03/01/2024] Open
Abstract
BACKGROUND Past studies have demonstrated that patients diagnosed with rheumatoid arthritis (RA) often exhibit abnormal levels of lipids. Furthermore, certain lipid-modifying medications have shown effectiveness in alleviating clinical symptoms associated with RA. However, the current understanding of the causal relationship between lipids, lipid-modifying medications, and the risk of developing RA remains inconclusive. This study employed Mendelian randomization (MR) to investigate the causal connection between lipids, lipid-modifying drugs, and the occurrence of RA. METHODS We obtained genetic variation for lipid traits and drug targets related to lipid modification from three sources: the Global Lipids Genetics Consortium (GLGC), UK Biobank, and Nightingale Health 2020. The genetic data for RA were acquired from two comprehensive meta-analyses and the R8 of FINNGEN, respectively. These variants were employed in drug-target MR analyses to establish a causal relationship between genetically predicted lipid-modifying drug targets and the risk of RA. For suggestive lipid-modified drug targets, we conducted Summary-data-based Mendelian Randomization (SMR) analyses and using expression quantitative trait loci (eQTL) data in relevant tissues. In addition, we performed co-localization analyses to assess genetic confounders. RESULTS Our analysis revealed no significant causal relationship between lipid and RA. We observed that the genetically predicted 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) -mediated low density lipoprotein cholesterol (LDL-C) (OR 0.704; 95% CI 0.56, 0.89; P = 3.43×10-3), Apolipoprotein C-III (APOC3) -mediated triglyceride (TG) (OR 0.844; 95% CI 0.77, 0.92; P = 1.50×10-4) and low density lipoprotein receptor (LDLR) -mediated LDL-C (OR 0.835; 95% CI 0.73, 0.95; P = 8.81×10-3) were significantly associated with a lowered risk of RA. while Apolipoprotein B-100 (APOB) -mediated LDL-C (OR 1.212; 95%CI 1.05,1.40; P = 9.66×10-3) was significantly associated with an increased risk of RA. CONCLUSIONS Our study did not find any supporting evidence to suggest that lipids are a risk factor for RA. However, we observed significant associations between HMGCR, APOC3, LDLR, and APOB with the risk of RA.
Collapse
Affiliation(s)
- Zhican Huang
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ting Cui
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jin Yao
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yutong Wu
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Zhu
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xin Yang
- School of Health and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Cui
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Haiyan Zhou
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
223
|
Lin L, Kiryakos J, Ammous F, Ratliff SM, Ware EB, Faul JD, Kardia SLR, Zhao W, Birditt KS, Smith JA. Epigenetic age acceleration is associated with blood lipid levels in a multi-ancestry sample of older U.S. adults. RESEARCH SQUARE 2024:rs.3.rs-3934965. [PMID: 38464171 PMCID: PMC10925395 DOI: 10.21203/rs.3.rs-3934965/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Background Dyslipidemia, which is characterized by an unfavorable lipid profile, is a key risk factor for cardiovascular disease (CVD). Understanding the relationships between epigenetic aging and lipid levels may help guide early prevention and treatment efforts for dyslipidemia. Methods We used weighted linear regression to cross-sectionally investigate the associations between five measures of epigenetic age acceleration estimated from whole blood DNA methylation (HorvathAge Acceleration, HannumAge Acceleration, PhenoAge Acceleration, GrimAge Acceleration, and DunedinPACE) and four blood lipid measures (total cholesterol (TC), LDL-C, HDL-C, and triglycerides (TG)) in 3,813 participants (mean age = 70 years) from the Health and Retirement Study (HRS). As a sensitivity analysis, we examined the same associations in participants who fasted prior to the blood draw (n = and f) and in participants who did not take lipid-lowering medication (n = 1,869). Using interaction models, we also examined whether the relationships between epigenetic age acceleration and blood lipids differ by demographic factors including age, sex, and educational attainment. Results After adjusting for age, race/ethnicity, sex, fasting status, and lipid-lowering medication use, greater epigenetic age acceleration was associated with lower TC, HDL-C, and LDL-C, and higher TG (p < 0.05). GrimAge acceleration and DunedinPACE associations with all lipids remained significant after further adjusting for body mass index, smoking status, and educational attainment. These associations were stronger in participants who fasted and who did not use lipid-lowering medication, particularly for LDL-C. We observed the largest number of interactions between DunedinPACE and demographic factors, where the associations with lipids were stronger in younger participants, females, and those with higher educational attainment. Conclusion Epigenetic age acceleration, a powerful biomarker of cellular aging, is highly associated with blood lipid levels in older adults. A greater understanding of how these associations differ across demographic groups can help shed light on the relationships between aging and downstream cardiovascular diseases. The inverse associations between epigenetic age and TC and LDL-C could be due to sample limitations or the non-linear relationship between age and these lipids, as both TC and LDL-C decrease faster at older ages. More studies are needed to further understand the temporal relationships between epigenetic age acceleration on blood lipids and other health outcomes.
Collapse
Affiliation(s)
- Lisha Lin
- Department of Epidemiology, School of Public Health, University of Michigan
| | - Jenna Kiryakos
- Department of Epidemiology, School of Public Health, University of Michigan
| | - Farah Ammous
- Department of Epidemiology, School of Public Health, University of Michigan
| | - Scott M Ratliff
- Department of Epidemiology, School of Public Health, University of Michigan
| | - Erin B Ware
- Survey Research Center, Institute for Social Research, University of Michigan
| | - Jessica D Faul
- Survey Research Center, Institute for Social Research, University of Michigan
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan
| | - Kira S Birditt
- Survey Research Center, Institute for Social Research, University of Michigan
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan
| |
Collapse
|
224
|
Reshetnikov E, Churnosova M, Reshetnikova Y, Stepanov V, Bocharova A, Serebrova V, Trifonova E, Ponomarenko I, Sorokina I, Efremova O, Orlova V, Batlutskaya I, Ponomarenko M, Churnosov V, Aristova I, Polonikov A, Churnosov M. Maternal Age at Menarche Genes Determines Fetal Growth Restriction Risk. Int J Mol Sci 2024; 25:2647. [PMID: 38473894 PMCID: PMC10932237 DOI: 10.3390/ijms25052647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/06/2024] [Accepted: 02/14/2024] [Indexed: 03/14/2024] Open
Abstract
We aimed to explore the potential link of maternal age at menarche (mAAM) gene polymorphisms with risk of the fetal growth restriction (FGR). This case (FGR)-control (FGR free) study included 904 women (273 FGR and 631 control) in the third trimester of gestation examined/treated in the Departments of Obstetrics. For single nucleotide polymorphism (SNP) multiplex genotyping, 50 candidate loci of mAAM were chosen. The relationship of mAAM SNPs and FGR was appreciated by regression procedures (logistic/model-based multifactor dimensionality reduction [MB-MDR]) with subsequent in silico assessment of the assumed functionality pithy of FGR-related loci. Three mAAM-appertain loci were FGR-linked to genes such as KISS1 (rs7538038) (effect allele G-odds ratio (OR)allelic = 0.63/pperm = 0.0003; ORadditive = 0.61/pperm = 0.001; ORdominant = 0.56/pperm = 0.001), NKX2-1 (rs999460) (effect allele A-ORallelic = 1.37/pperm = 0.003; ORadditive = 1.45/pperm = 0.002; ORrecessive = 2.41/pperm = 0.0002), GPRC5B (rs12444979) (effect allele T-ORallelic = 1.67/pperm = 0.0003; ORdominant = 1.59/pperm = 0.011; ORadditive = 1.56/pperm = 0.009). The haplotype ACA FSHB gene (rs555621*rs11031010*rs1782507) was FRG-correlated (OR = 0.71/pperm = 0.05). Ten FGR-implicated interworking models were founded for 13 SNPs (pperm ≤ 0.001). The rs999460 NKX2-1 and rs12444979 GPRC5B interplays significantly influenced the FGR risk (these SNPs were present in 50% of models). FGR-related mAAM-appertain 15 polymorphic variants and 350 linked SNPs were functionally momentous in relation to 39 genes participating in the regulation of hormone levels, the ovulation cycle process, male gonad development and vitamin D metabolism. Thus, this study showed, for the first time, that the mAAM-appertain genes determine FGR risk.
Collapse
Affiliation(s)
- Evgeny Reshetnikov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
| | - Maria Churnosova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
| | - Yuliya Reshetnikova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
| | - Vadim Stepanov
- Research Institute for Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634050 Tomsk, Russia; (V.S.); (A.B.); (V.S.); (E.T.)
| | - Anna Bocharova
- Research Institute for Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634050 Tomsk, Russia; (V.S.); (A.B.); (V.S.); (E.T.)
| | - Victoria Serebrova
- Research Institute for Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634050 Tomsk, Russia; (V.S.); (A.B.); (V.S.); (E.T.)
| | - Ekaterina Trifonova
- Research Institute for Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, 634050 Tomsk, Russia; (V.S.); (A.B.); (V.S.); (E.T.)
| | - Irina Ponomarenko
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
| | - Inna Sorokina
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
| | - Olga Efremova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
| | - Valentina Orlova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
| | - Irina Batlutskaya
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
| | - Marina Ponomarenko
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
| | - Vladimir Churnosov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
| | - Inna Aristova
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
| | - Alexey Polonikov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
- Department of Biology, Medical Genetics and Ecology and Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 305041 Kursk, Russia
| | - Mikhail Churnosov
- Department of Medical Biological Disciplines, Belgorod State National Research University, 308015 Belgorod, Russia; (E.R.); (M.C.); (Y.R.); (I.P.); (I.S.); (O.E.); (V.O.); (I.B.); (M.P.); (V.C.); (I.A.); (A.P.)
| |
Collapse
|
225
|
Yan Z, Xu Y, Li K, Liu L. Association between high-density lipoprotein cholesterol and type 2 diabetes mellitus: dual evidence from NHANES database and Mendelian randomization analysis. Front Endocrinol (Lausanne) 2024; 15:1272314. [PMID: 38455653 PMCID: PMC10917910 DOI: 10.3389/fendo.2024.1272314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 02/12/2024] [Indexed: 03/09/2024] Open
Abstract
Background Low levels of high-density lipoprotein cholesterol (HDL-C) are commonly seen in patients with type 2 diabetes mellitus (T2DM). However, it is unclear whether there is an independent or causal link between HDL-C levels and T2DM. This study aims to address this gap by using the The National Health and Nutrition Examination Survey (NHANES) database and Mendelian randomization (MR) analysis. Materials and methods Data from the NHANES survey (2007-2018) with 9,420 participants were analyzed using specialized software. Logistic regression models and restricted cubic splines (RCS) were used to assess the relationship between HDL-C and T2DM incidence, while considering covariates. Genetic variants associated with HDL-C and T2DM were obtained from genome-wide association studies (GWAS), and Mendelian randomization (MR) was used to evaluate the causal relationship between HDL-C and T2DM. Various tests were conducted to assess pleiotropy and outliers. Results In the NHANES study, all groups, except the lowest quartile (Q1: 0.28-1.09 mmol/L], showed a significant association between HDL-C levels and reduced T2DM risk (all P < 0.001). After adjusting for covariates, the Q2 [odds ratio (OR) = 0.67, 95% confidence interval (CI): (0.57, 0.79)], Q3 [OR = 0.51, 95% CI: (0.40, 0.65)], and Q4 [OR = 0.29, 95% CI: (0.23, 0.36)] groups exhibited average reductions in T2DM risk of 23%, 49%, and 71%, respectively. In the sensitivity analysis incorporating other lipid levels, the Q4 group still demonstrates a 57% reduction in the risk of T2DM. The impact of HDL-C levels on T2DM varied with age (P for interaction = 0.006). RCS analysis showed a nonlinear decreasing trend in T2DM risk with increasing HDL-C levels (P = 0.003). In the MR analysis, HDL-C levels were also associated with reduced T2DM risk (OR = 0.69, 95% CI = 0.52-0.82; P = 1.41 × 10-13), and there was no evidence of pleiotropy or outliers. Conclusion This study provides evidence supporting a causal relationship between higher HDL-C levels and reduced T2DM risk. Further research is needed to explore interventions targeting HDL-C levels for reducing T2DM risk.
Collapse
Affiliation(s)
- Zhaoqi Yan
- Jiangxi University of Traditional Chinese Medicine, Graduate School, Nanchang, Jiangxi, China
| | - Yifeng Xu
- Jiangxi University of Traditional Chinese Medicine, Graduate School, Nanchang, Jiangxi, China
| | - Keke Li
- Jiangxi University of Traditional Chinese Medicine, Graduate School, Nanchang, Jiangxi, China
| | - Liangji Liu
- Affiliated Hospital of Jiangxi University of Traditional Chinese Medicine, Department of Respiratory and Critical Care Medicine, Nanchang, Jiangxi, China
| |
Collapse
|
226
|
Martin SS, Aday AW, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Barone Gibbs B, Beaton AZ, Boehme AK, Commodore-Mensah Y, Currie ME, Elkind MSV, Evenson KR, Generoso G, Heard DG, Hiremath S, Johansen MC, Kalani R, Kazi DS, Ko D, Liu J, Magnani JW, Michos ED, Mussolino ME, Navaneethan SD, Parikh NI, Perman SM, Poudel R, Rezk-Hanna M, Roth GA, Shah NS, St-Onge MP, Thacker EL, Tsao CW, Urbut SM, Van Spall HGC, Voeks JH, Wang NY, Wong ND, Wong SS, Yaffe K, Palaniappan LP. 2024 Heart Disease and Stroke Statistics: A Report of US and Global Data From the American Heart Association. Circulation 2024; 149:e347-e913. [PMID: 38264914 DOI: 10.1161/cir.0000000000001209] [Citation(s) in RCA: 659] [Impact Index Per Article: 659.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
BACKGROUND The American Heart Association (AHA), in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, nutrition, sleep, and obesity) and health factors (cholesterol, blood pressure, glucose control, and metabolic syndrome) that contribute to cardiovascular health. The AHA Heart Disease and Stroke Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, brain health, complications of pregnancy, kidney disease, congenital heart disease, rhythm disorders, sudden cardiac arrest, subclinical atherosclerosis, coronary heart disease, cardiomyopathy, heart failure, valvular disease, venous thromboembolism, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The AHA, through its Epidemiology and Prevention Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States and globally to provide the most current information available in the annual Statistical Update with review of published literature through the year before writing. The 2024 AHA Statistical Update is the product of a full year's worth of effort in 2023 by dedicated volunteer clinicians and scientists, committed government professionals, and AHA staff members. The AHA strives to further understand and help heal health problems inflicted by structural racism, a public health crisis that can significantly damage physical and mental health and perpetuate disparities in access to health care, education, income, housing, and several other factors vital to healthy lives. This year's edition includes additional global data, as well as data on the monitoring and benefits of cardiovascular health in the population, with an enhanced focus on health equity across several key domains. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
227
|
Reay WR, Kiltschewskij DJ, Di Biase MA, Gerring ZF, Kundu K, Surendran P, Greco LA, Clarke ED, Collins CE, Mondul AM, Albanes D, Cairns MJ. Genetic influences on circulating retinol and its relationship to human health. Nat Commun 2024; 15:1490. [PMID: 38374065 PMCID: PMC10876955 DOI: 10.1038/s41467-024-45779-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 02/04/2024] [Indexed: 02/21/2024] Open
Abstract
Retinol is a fat-soluble vitamin that plays an essential role in many biological processes throughout the human lifespan. Here, we perform the largest genome-wide association study (GWAS) of retinol to date in up to 22,274 participants. We identify eight common variant loci associated with retinol, as well as a rare-variant signal. An integrative gene prioritisation pipeline supports novel retinol-associated genes outside of the main retinol transport complex (RBP4:TTR) related to lipid biology, energy homoeostasis, and endocrine signalling. Genetic proxies of circulating retinol were then used to estimate causal relationships with almost 20,000 clinical phenotypes via a phenome-wide Mendelian randomisation study (MR-pheWAS). The MR-pheWAS suggests that retinol may exert causal effects on inflammation, adiposity, ocular measures, the microbiome, and MRI-derived brain phenotypes, amongst several others. Conversely, circulating retinol may be causally influenced by factors including lipids and serum creatinine. Finally, we demonstrate how a retinol polygenic score could identify individuals more likely to fall outside of the normative range of circulating retinol for a given age. In summary, this study provides a comprehensive evaluation of the genetics of circulating retinol, as well as revealing traits which should be prioritised for further investigation with respect to retinol related therapies or nutritional intervention.
Collapse
Affiliation(s)
- William R Reay
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia.
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia.
| | - Dylan J Kiltschewskij
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Maria A Di Biase
- Melbourne Neuropsychiatry Centre, Department of Psychiatry, The University of Melbourne, Melbourne, VIC, Australia
- Department of Anatomy and Physiology, The University of Melbourne, Melbourne, VIC, Australia
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Zachary F Gerring
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Kousik Kundu
- Human Genetics, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Praveen Surendran
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- British Heart Foundation Centre of Research Excellence, School of Clinical Medicine, Addenbrooke's Hospital, University of Cambridge, Cambridge, UK
- Health Data Research UK, Wellcome Genome Campus and University of Cambridge, Hinxton, UK
| | - Laura A Greco
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Erin D Clarke
- School of Health Sciences, The University of Newcastle, Callaghan, NSW, Australia
- Food and Nutrition Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Clare E Collins
- School of Health Sciences, The University of Newcastle, Callaghan, NSW, Australia
- Food and Nutrition Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia
| | - Alison M Mondul
- Department of Epidemiology, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Demetrius Albanes
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Department of Health and Human Services, Bethesda, MD, USA
| | - Murray J Cairns
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, NSW, Australia.
- Precision Medicine Research Program, Hunter Medical Research Institute, New Lambton, NSW, Australia.
| |
Collapse
|
228
|
Huang A, Wu X, Lin J, Wei C, Xu W. Genetic insights into repurposing statins for hyperthyroidism prevention: a drug-target Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1331031. [PMID: 38425755 PMCID: PMC10902122 DOI: 10.3389/fendo.2024.1331031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/01/2024] [Indexed: 03/02/2024] Open
Abstract
Background Current therapeutic measures for thyroid dysfunction are limited and often accompanied by adverse effects. The use of lipid-lowering drugs like statins has recently been associated with lower thyroid eye diseases risk. Objective To investigate the implications of genetically proxied lipid-lowering drugs on thyroid dysfunction. Methods In this drug-target Mendelian randomization (MR) study, we utilized genetic variants within drug target genes associated with low-density lipoprotein (LDL) or triglyceride (TG), derived from a genome-wide association study (GWAS) meta-analysis (N ≤ 188,577), to simulate lifelong drug interventions. Genetic summary statistics for thyroid dysfunction outcomes were retrieved from GWAS datasets of Thyroid Omics Consortium (N ≤ 54,288) and UK Biobank (N = 484,598). Inverse-variance-weighted MR (IVW-MR) method was performed as primary analysis, followed by validation in colocalization analysis. A subsequent two-step MR analysis was conducted to identify biomarkers mediating the identified drug-outcome association. Results In IVW-MR analysis, genetic mimicry of 3-hydroxy-3-methylglutarylcoenzyme reductase (HMGCR) inhibitors (e.g. statins) was significantly associated with lower risk of hyperthyroidism in two independent datasets (OR1, 0.417 per 1-mmol/L lower in LDL-C; 95% CI 0.262 to 0.664; P1 = 2.262 × 10-4; OR2 0.996; 95% CI 0.993-0.998; P2 = 0.002). Two-step MR analysis revealed eighteen biomarkers linked to genetic mimicry of HMGCR inhibition, and identified insulin-like growth factor 1 (IGF-1) levels mediating 2.108% of the negative causal relationship between HMGCR inhibition and hyperthyroidism. Conclusion This study supports HMGCR inhibition as a promising therapeutic strategy for hyperthyroidism and suggests its underlying mechanisms may extend beyond lipid metabolism. Further investigations through laboratory studies and clinical trials are necessary to confirm and elucidate these findings.
Collapse
Affiliation(s)
- Anqi Huang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Shantou University Medical College, Shantou, China
| | - Xinyi Wu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Shantou University Medical College, Shantou, China
| | - Jiaqi Lin
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
- Shantou University Medical College, Shantou, China
| | - Chiju Wei
- Multidisciplinary Research Center, Shantou University, Shantou, China
| | - Wencan Xu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| |
Collapse
|
229
|
Hu M, Yang T, Yang Y. Causal Associations of Education Level With Cardiovascular Diseases, Cardiovascular Biomarkers, and Socioeconomic Factors. Am J Cardiol 2024; 213:76-85. [PMID: 38199144 DOI: 10.1016/j.amjcard.2023.06.044] [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: 03/29/2023] [Revised: 05/12/2023] [Accepted: 06/11/2023] [Indexed: 01/12/2024]
Abstract
An inverse association of education level with cardiovascular diseases has been documented in observational studies, yet the causality and potential mechanisms remain to be determined. To systematically investigate the causal associations of education level with cardiovascular diseases, cardiovascular biomarkers, and socioeconomic factors, a 2-sample Mendelian randomization was performed. The results revealed that higher genetically determined education level was associated with lower risks of type 2 diabetes mellitus (odds ratio [OR] 0.54, 95% confidence interval [CI] 0.47 to 0.61, p = 3.04 × 10-23), peripheral artery disease (OR 0.62, 95% CI 0.51 to 0.76, p = 2.14 × 10-06), hypertension (OR 0.62, 95% CI 0.56 to 0.70, p = 4.22 × 10-16), coronary heart disease (OR 0.62, 95% CI 0.56 to 0.69, p = 3.50 × 10-19), myocardial infarction (OR 0.62, 95% CI 0.55 to 0.69, p = 2.58 × 10-16), ischemic stroke (OR 0.67, 95% CI 0.62 to 0.74, p = 6.00 × 10-19), deep vein thrombosis (OR 0.69, 95% CI 0.55 to 0.87, p = 0.0017), atrial fibrillation (OR 0.70, 95% CI 0.57 to 0.86, p = 0.0007), cardiac death (OR 0.71, 95% CI 0.60 to 0.86, p = 0.0003), heart failure (OR 0.72, 95% CI 0.65 to 0.79, p = 6.37 × 10-12), transient ischemic attack (OR 0.76, 95% CI 0.64 to 0.90, p = 0.0010), and venous thromboembolism (OR 0.79, 95% CI 0.67 to 0.92, p = 0.0028). Systolic blood pressure, diastolic blood pressure, C-reactive protein, body mass index, waist circumference, and triglycerides were decreased, whereas telomere length was increased. Subjects with higher education were less likely to smoke, intake salt, or be exposed to air pollution and depression state. They were more likely to take physical activity and possess more household income. In conclusion, higher education may causally decrease cardiovascular diseases through socioeconomic factors and cardiovascular biomarkers. Reducing education inequality is important in the management of cardiovascular diseases.
Collapse
Affiliation(s)
- Mengjin Hu
- Department of Cardiology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tao Yang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuejin Yang
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
230
|
Arunachalam V, Lea R, Hoy W, Lee S, Mott S, Savige J, Mathews JD, McMorran BJ, Nagaraj SH. Novel genetic markers for chronic kidney disease in a geographically isolated population of Indigenous Australians: Individual and multiple phenotype genome-wide association study. Genome Med 2024; 16:29. [PMID: 38347632 PMCID: PMC10860247 DOI: 10.1186/s13073-024-01299-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 01/30/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) is highly prevalent among Indigenous Australians, especially those in remote regions. The Tiwi population has been isolated from mainland Australia for millennia and exhibits unique genetic characteristics that distinguish them from other Indigenous and non-Indigenous populations. Notably, the rate of end-stage renal disease is up to 20 times greater in this population compared to non-Indigenous populations. Despite the identification of numerous genetic loci associated with kidney disease through GWAS, the Indigenous population such as Tiwi remains severely underrepresented and the increased prevalence of CKD in this population may be due to unique disease-causing alleles/genes. METHODS We used albumin-to-creatinine ratio (ACR) and estimated glomerular filtration rate (eGFR) to estimate the prevalence of kidney disease in the Tiwi population (N = 492) in comparison to the UK Biobank (UKBB) (N = 134,724) database. We then performed an exploratory factor analysis to identify correlations among 10 CKD-related phenotypes and identify new multi-phenotype factors. We subsequently conducted a genome-wide association study (GWAS) on all single and multiple phenotype factors using mixed linear regression models, adjusted for age, sex, population stratification, and genetic relatedness between individuals. RESULTS Based on ACR, 20.3% of the population was at severely increased risk of CKD progression and showed elevated levels of ACR compared to the UKBB population independent of HbA1c. A GWAS of ACR revealed novel association loci in the genes MEG3 (chr14:100812018:T:A), RAB36 (rs11704318), and TIAM2 (rs9689640). Additionally, multiple phenotypes GWAS of ACR, eGFR, urine albumin, and serum creatinine identified a novel variant that mapped to the gene MEIS2 (chr15:37218869:A:G). Most of the identified variants were found to be either absent or rare in the UKBB population. CONCLUSIONS Our study highlights the Tiwi population's predisposition towards elevated ACR, and the collection of novel genetic variants associated with kidney function. These associations may prove valuable in the early diagnosis and treatment of renal disease in this underrepresented population. Additionally, further research is needed to comprehensively validate the functions of the identified variants/genes.
Collapse
Affiliation(s)
- Vignesh Arunachalam
- Centre for Genomics and Personalised Health and School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Rodney Lea
- Centre for Genomics and Personalised Health and School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Wendy Hoy
- Centre of chronic disease, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Simon Lee
- Centre for Genomics and Personalised Health and School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Susan Mott
- Centre of chronic disease, Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Judith Savige
- Royal Melbourne Hospital, The University of Melbourne, Melbourne, VIC, Australia
| | - John D Mathews
- Melbourne School of Population and Global Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Brendan J McMorran
- National Centre for Indigenous Genomics, The John Curtin of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Shivashankar H Nagaraj
- Centre for Genomics and Personalised Health and School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.
- Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia.
| |
Collapse
|
231
|
Zhuang Y, Kim NY, Fritsche LG, Mukherjee B, Lee S. Incorporating functional annotation with bilevel continuous shrinkage for polygenic risk prediction. BMC Bioinformatics 2024; 25:65. [PMID: 38336614 PMCID: PMC11323637 DOI: 10.1186/s12859-024-05664-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Genetic variants can contribute differently to trait heritability by their functional categories, and recent studies have shown that incorporating functional annotation can improve the predictive performance of polygenic risk scores (PRSs). In addition, when only a small proportion of variants are causal variants, PRS methods that employ a Bayesian framework with shrinkage can account for such sparsity. It is possible that the annotation group level effect is also sparse. However, the number of PRS methods that incorporate both annotation information and shrinkage on effect sizes is limited. We propose a PRS method, PRSbils, which utilizes the functional annotation information with a bilevel continuous shrinkage prior to accommodate the varying genetic architectures both on the variant-specific level and on the functional annotation level. RESULTS We conducted simulation studies and investigated the predictive performance in settings with different genetic architectures. Results indicated that when there was a relatively large variability of group-wise heritability contribution, the gain in prediction performance from the proposed method was on average 8.0% higher AUC compared to the benchmark method PRS-CS. The proposed method also yielded higher predictive performance compared to PRS-CS in settings with different overlapping patterns of annotation groups and obtained on average 6.4% higher AUC. We applied PRSbils to binary and quantitative traits in three real world data sources (the UK Biobank, the Michigan Genomics Initiative (MGI), and the Korean Genome and Epidemiology Study (KoGES)), and two sources of annotations: ANNOVAR, and pathway information from the Kyoto Encyclopedia of Genes and Genomes (KEGG), and demonstrated that the proposed method holds the potential for improving predictive performance by incorporating functional annotations. CONCLUSIONS By utilizing a bilevel shrinkage framework, PRSbils enables the incorporation of both overlapping and non-overlapping annotations into PRS construction to improve the performance of genetic risk prediction. The software is available at https://github.com/styvon/PRSbils .
Collapse
Affiliation(s)
| | - Na Yeon Kim
- Seoul National University, Seoul, Republic of Korea
| | | | | | - Seunggeun Lee
- Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
232
|
Gao B, Zhou Z, Chen J, Zhang S, Jin S, Yang W, Lei Y, Wang K, Li J, Zhuang Y. Aminopeptidase O Protein mediates the association between Lachnospiraceae and appendicular lean mass. Front Microbiol 2024; 15:1325466. [PMID: 38384268 PMCID: PMC10879621 DOI: 10.3389/fmicb.2024.1325466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 01/25/2024] [Indexed: 02/23/2024] Open
Abstract
Objective Investigating the causal relationship between Lachnospiraceae and Appendicular lean mass (ALM) and identifying and quantifying the role of Aminopeptidase O Protein (AOPEP) as a potential mediator. Methods The summary statistics data of gut microbiota composition from the largest available genome-wide association study (GWAS) meta-analysis conducted by the MiBioGen Consortium (n = 13,266). Appendicular lean mass data were obtained from the UK-Biobank (n = 450,243). We conducted bidirectional two-sample Mendelian randomization (MR) analysis using summary-level data from GWAS to investigate the causal relationship between Lachnospiraceae and ALM. Additionally, we employed a drug-targeted MR approach to assess the causal relationship between AOPEP and ALM. Finally, a two-step MR was employed to quantitatively estimate the proportion of the effect of Lachnospiraceae on ALM that is mediated by AOPEP. Cochran's Q statistic was used to quantify heterogeneity among instrumental variable estimates. Results In the MR analysis, it was found that an increase in genetically predicted Lachnospiraceae [OR = 1.031, 95% CI (1.011-1.051), P = 0.002] is associated with an increase in ALM. There is no strong evidence to suggest that genetically predicted ALM has an impact on Lachnospiraceae genus [OR = 1.437, 95% CI (0.785-2.269), P = 0.239]. The proportion of genetically predicted Lachnospiraceae mediated by AOPEP was 34.2% [95% CI (1.3%-67.1%)]. Conclusion Our research reveals that increasing Lachnospiraceae abundance in the gut can directly enhance limb muscle mass and concurrently suppress AOPEP, consequently mitigating limb muscle loss. This supports the potential therapeutic modulation of gut microbiota for sarcopenia. Interventions such as drug treatments or microbiota transplantation, aimed at elevating Lachnospiraceae abundance and AOPEP inhibition, synergistically improve sarcopenia in the elderly, thereby enhancing the overall quality of life for older individuals.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yan Zhuang
- Department of Pediatric Surgery, Qilu Hospital of Shandong University, Jinan, Shandong, China
| |
Collapse
|
233
|
Zhang Y, Tang Z, Shi Y, Li L. Associations between artificial sweetener intake from cereals, coffee, and tea and the risk of type 2 diabetes mellitus: A genetic correlation, mediation, and mendelian randomization analysis. PLoS One 2024; 19:e0287496. [PMID: 38324548 PMCID: PMC10849235 DOI: 10.1371/journal.pone.0287496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 10/13/2023] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND Previous studies have emphasized the association between the intake of artificial sweeteners (AS) and type 2 diabetes mellitus (T2DM), but the causative relationship remains ambiguous. METHODS This study employed univariate Mendelian randomization (MR) analysis to assess the causal link between AS intake from various sources and T2DM. Linkage disequilibrium score (LDSC) regression was used to evaluate the correlation between phenotypes. Multivariate and mediation MR were applied to investigate confounding factors and mediating effects. Data on AS intake from different sources (N = 64,949) were sourced from the UK Biobank, while T2DM data were derived from the DIAbetes Genetics Replication And Meta-analysis.The primary method adopted was inverse variance weighted (IVW), complemented by three validation techniques. Additionally, a series of sensitivity analyses were performed to evaluate pleiotropy and heterogeneity. RESULTS LDSC analysis unveiled a significant genetic correlation between AS intake from different sources and T2DM (rg range: -0.006 to 0.15, all P < 0.05). After correction by the false discovery rate (FDR), the primary IVW method indicated that AS intake in coffee was a risk factor for T2DM (OR = 1.265, 95% CI: 1.035-1.545, P = 0.021, PFDR = 0.042). Further multivariable and mediation MR analyses pinpointed high density lipoprotein-cholesterol (HDL-C) as mediating a portion of this causal relationship. In reverse MR analysis, significant evidence suggested a positive correlation between T2DM and AS intake in coffee (β = 0.013, 95% CI: 0.004-0.022, P = 0.004, PFDR = 0.012), cereal (β = 0.007, 95% CI: 0.002-0.012, P = 0.004, PFDR = 0.012), and tea (β = 0.009, 95% CI: 0.001-0.017, P = 0.036, PFDR = 0.049). No other causal associations were identified (P > 0.05, PFDR > 0.05). CONCLUSION The MR analysis has established a causal relationship between AS intake in coffee and T2DM. The mediation by HDL-C emphasizes potential metabolic pathways underpinning these relationships.
Collapse
Affiliation(s)
- Youqian Zhang
- Department of Endocrinology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| | - Zitian Tang
- Department of Law, Yangtze University, Jingzhou, Hubei, China
| | - Yong Shi
- Department of Medicine, Yangtze University, Jingzhou, Hubei, China
| | - Lin Li
- Department of Endocrinology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei, China
| |
Collapse
|
234
|
Salih AM, Galazzo IB, Menegaz G, Altmann A. Leukocyte Telomere Length and Cardiac Structure and Function: A Mendelian Randomization Study. J Am Heart Assoc 2024; 13:e032708. [PMID: 38293941 PMCID: PMC11056120 DOI: 10.1161/jaha.123.032708] [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: 10/30/2023] [Accepted: 01/04/2024] [Indexed: 02/01/2024]
Abstract
BACKGROUND Existing research demonstrates the association of shorter leukocyte telomere length with increased risk of age-related health outcomes including cardiovascular diseases. However, the direct causality of these relationships has not been definitively established. Cardiovascular aging at an organ level may be captured using image-derived phenotypes of cardiac anatomy and function. METHODS AND RESULTS In the current study, we use 2-sample Mendelian randomization to assess the causal link between leukocyte telomere length and 54 cardiac magnetic resonance imaging measures representing structure and function across the 4 cardiac chambers. Genetically predicted shorter leukocyte telomere length was causally linked to smaller ventricular cavity sizes including left ventricular end-systolic volume, left ventricular end-diastolic volume, lower left ventricular mass, and pulmonary artery. The association with left ventricular mass (β =0.217, Pfalse discovery rate=0.016) remained significant after multiple testing adjustment, whereas other associations were attenuated. CONCLUSIONS Our findings support a causal role for shorter leukocyte telomere length and faster cardiac aging, with the most prominent relationship with left ventricular mass.
Collapse
Affiliation(s)
- Ahmed M. Salih
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University of LondonUK
- Department of Population Health SciencesUniversity of LeicesterUK
- Department of Computer ScienceUniversity of ZakhoKurdistan of IraqIraq
| | | | | | - André Altmann
- Centre for Medical Image Computing, Department of Medical Physics and Biomedical EngineeringUniversity College LondonUK
| |
Collapse
|
235
|
Kim SJ, Miller B, Hartel NG, Ramirez R, Braniff RG, Leelaprachakul N, Huang A, Wang Y, Arpawong TE, Crimmins EM, Wang P, Sun X, Liu C, Levy D, Yen K, Petzinger GM, Graham NA, Jakowec MW, Cohen P. A naturally occurring variant of SHLP2 is a protective factor in Parkinson's disease. Mol Psychiatry 2024; 29:505-517. [PMID: 38167865 PMCID: PMC11116102 DOI: 10.1038/s41380-023-02344-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Revised: 11/20/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024]
Abstract
Mitochondrial DNA single nucleotide polymorphisms (mtSNPs) have been associated with a reduced risk of developing Parkinson's disease (PD), yet the underlying mechanisms remain elusive. In this study, we investigate the functional role of a PD-associated mtSNP that impacts the mitochondrial-derived peptide (MDP) Small Humanin-like Peptide 2 (SHLP2). We identify m.2158 T > C, a mtSNP associated with reduced PD risk, within the small open reading frame encoding SHLP2. This mtSNP results in an alternative form of SHLP2 (lysine 4 replaced with arginine; K4R). Using targeted mass spectrometry, we detect specific tryptic fragments of SHLP2 in neuronal cells and demonstrate its binding to mitochondrial complex 1. Notably, we observe that the K4R variant, associated with reduced PD risk, exhibits increased stability compared to WT SHLP2. Additionally, both WT and K4R SHLP2 show enhanced protection against mitochondrial dysfunction in in vitro experiments and confer protection against a PD-inducing toxin, a mitochondrial complex 1 inhibitor, in a mouse model. This study sheds light on the functional consequences of the m.2158 T > C mtSNP on SHLP2 and provides insights into the potential mechanisms by which this mtSNP may reduce the risk of PD.
Collapse
Affiliation(s)
- Su-Jeong Kim
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Brendan Miller
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Nicolas G Hartel
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, USA
| | - Ricardo Ramirez
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Regina Gonzalez Braniff
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Naphada Leelaprachakul
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Environmental Toxicology Program, Chulabhorn Graduate Institute, Bangkok, 10210, Thailand
| | - Amy Huang
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Yuzhu Wang
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Thalida Em Arpawong
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Eileen M Crimmins
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Penglong Wang
- The Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xianbang Sun
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Chunyu Liu
- The Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Boston University's and National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA, USA
| | - Daniel Levy
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Boston University's and National Heart, Lung, and Blood Institute's Framingham Heart Study, Framingham, MA, USA
| | - Kelvin Yen
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Giselle M Petzinger
- Department of Neurology, University of Southern California, Los Angeles, CA, USA
| | - Nicholas A Graham
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA, USA
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, CA, USA
| | - Michael W Jakowec
- Department of Neurology, University of Southern California, Los Angeles, CA, USA
- Department of Biokinesiology and Physical Therapy, The George and MaryLou Boone Center for Parkinson's Disease Research, University of Southern California, Los Angeles, CA, USA
| | - Pinchas Cohen
- The Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
236
|
Zhang Z, Duan Y, Huo J. Lipid Metabolism, Methylation Aberrant, and Osteoporosis: A Multi-omics Study Based on Mendelian Randomization. Calcif Tissue Int 2024; 114:147-156. [PMID: 38071623 DOI: 10.1007/s00223-023-01160-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 11/03/2023] [Indexed: 01/23/2024]
Abstract
BACKGROUND Observational studies have shown a causal association between dyslipidemia and osteoporosis, but the genetic causation and complete mechanism of which are uncertain. The disadvantage of previous observational studies is that they are susceptible to confounding factors and bias, that makes it difficult to infer a causal link between those two diseases. Abnormal epigenetic modifications, represented by DNA methylation, are important causes of many diseases. However, there are no studies showing a bridging role for methylation modifications in blood lipid metabolism and osteoporosis. METHODS SNPs for lipid profile (Blood VLDL cholesterol (VLDL-C), blood LDL cholesterol (LDL-C), blood HDL cholesterol (HDL-C), blood triglycerides (TG), diagnosed pure hypercholesterolaemia, blood apolipoprotein B (Apo B), blood apolipoprotein A1(Apo A1)), and bone mineral density (BMD) in different body parts (Heel BMD, lumbar BMD, whole-body BMD, femoral neck BMD) were obtained from large meta-analyses of genome-wide association studies as instrumental variables for two-sample Mendelian randomization. Assessment of the genetic effects of lipid profile-associated methylation sites and bone mineral density was carried out using the summary-data-based Mendelian randomization (SMR) method. RESULTS Two-sample Mendelian randomization showed that there was a negative causal association between hypercholesterolaemia and heel BMD (p = 0.0103, OR = 0.4590), and total body BMD (p = 0.0002, OR = 0.2826). LDL-C had a negative causal association with heel BMD (p = 8.68E-05, OR = 0.9586). VLDL-C had a negative causal association with heel BMD (p = 0.035, OR = 0.9484), lumbar BMD (p = 0.0316, OR = 0.9356), and total body BMD (p = 0.0035, OR = 0.9484). HDL-C had a negative causal association with heel BMD (p = 1.25E-05, OR = 0.9548), lumbar BMD (p = 0.0129, OR = 0.9358), and total body BMD (p = 0.0399, OR = 0.9644). Apo B had a negative causal association with heel BMD (p = 0.0001, OR = 0.9647). Apo A1 had a negative causal association with heel BMD (p = 0.0132, OR = 0.9746) and lumbar BMD (p = 0.0058, OR = 0.9261). The p-values of all positive results corrected by the FDR method remained significant and sensitivity analysis showed that there was no horizontal pleiotropy in the results despite the heterogeneity in some results. SMR identified 3 methylation sites associated with lipid profiles in the presence of genetic effects on BMD: cg15707428(GREB1), cg16000331(SREBF2), cg14364472(NOTCH1). CONCLUSION Our study provides insights into the potential causal links and co-pathogenesis between dyslipidemia and osteoporosis. The genetic effects of dyslipidaemia on osteoporosis may be related to certain aberrant methylation genetic modifications.
Collapse
Affiliation(s)
- ZhaoLiang Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - YuChen Duan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - JianZhong Huo
- Taiyuan Central Hospital, Ninth Hospital of Shanxi Medical University, Southern Fendong Road 256, Taiyuan, 030009, Shanxi, China.
| |
Collapse
|
237
|
Levi H, Elkon R, Shamir R. The predictive capacity of polygenic risk scores for disease risk is only moderately influenced by imputation panels tailored to the target population. Bioinformatics 2024; 40:btae036. [PMID: 38265251 PMCID: PMC10868313 DOI: 10.1093/bioinformatics/btae036] [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: 08/31/2023] [Revised: 12/20/2023] [Accepted: 01/20/2024] [Indexed: 01/25/2024] Open
Abstract
MOTIVATION Polygenic risk scores (PRSs) predict individuals' genetic risk of developing complex diseases. They summarize the effect of many variants discovered in genome-wide association studies (GWASs). However, to date, large GWASs exist primarily for the European population and the quality of PRS prediction declines when applied to other ethnicities. Genetic profiling of individuals in the discovery set (on which the GWAS was performed) and target set (on which the PRS is applied) is typically done by SNP arrays that genotype a fraction of common SNPs. Therefore, a key step in GWAS analysis and PRS calculation is imputing untyped SNPs using a panel of fully sequenced individuals. The imputation results depend on the ethnic composition of the imputation panel. Imputing genotypes with a panel of individuals of the same ethnicity as the genotyped individuals typically improves imputation accuracy. However, there has been no systematic investigation into the influence of the ethnic composition of imputation panels on the accuracy of PRS predictions when applied to ethnic groups that differ from the population used in the GWAS. RESULTS We estimated the effect of imputation of the target set on prediction accuracy of PRS when the discovery and the target sets come from different ethnic groups. We analyzed binary phenotypes on ethnically distinct sets from the UK Biobank and other resources. We generated ethnically homogenous panels, imputed the target sets, and generated PRSs. Then, we assessed the prediction accuracy obtained from each imputation panel. Our analysis indicates that using an imputation panel matched to the ethnicity of the target population yields only a marginal improvement and only under specific conditions. AVAILABILITY AND IMPLEMENTATION The source code used for executing the analyses is this paper is available at https://github.com/Shamir-Lab/PRS-imputation-panels.
Collapse
Affiliation(s)
- Hagai Levi
- The Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv 69978, Israel
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ran Elkon
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ron Shamir
- The Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
238
|
Dai Q, Guo Y, Dong X, Gao Y, Li S, Zhao J, Xu Y, Liu K. The role of leptin in ischemic stroke and its subtypes: A Mendelian randomization study. Nutr Metab Cardiovasc Dis 2024; 34:360-368. [PMID: 37949710 DOI: 10.1016/j.numecd.2023.09.001] [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: 04/30/2023] [Revised: 08/18/2023] [Accepted: 09/01/2023] [Indexed: 11/12/2023]
Abstract
BACKGROUND AND AIMS Observational studies have suggested a relationship between leptin and risk of stroke. However, evidence for the association remains inconsistent, and whether the association reflects a causal relationship remains to be established. To clarify this relationship, we adopted a two-sample Mendelian randomization (MR) analysis to investigate whether leptin plays a causal role in the risk of stroke and its subtypes. METHODS AND RESULTS Five independent single-nucleotide polymorphisms (SNPs) associated with the leptin level from genome-wide association studies (GWASs) of European individuals were selected. We performed an MR analysis using the inverse-variance-weighted (IVW) as primary method to examine the causal effects of leptin on ischemic stroke (IS). Moreover, MR-Egger intercept and Cochran's Q statistic were also performed to detect the pleiotropy or heterogeneity of our MR results. Genetically predicted circulating leptin level was not associated with ischemic stroke [odds ratio (OR): 1.48, 95% confidence interval (CI): 0.78-2.8, P = 0.22], large artery stroke (OR: 1.44, 95% CI: 0.39-5.25, P = 0.57), cardioembolic stroke (OR:1.33, 95% CI: 0.55-3.22, P = 0.52), and small vessel stroke (OR: 1.48, 95% CI: 0.39-5.63, P = 0.56) using the IVW method. Likewise, there is no convincing evidence for the associations between leptin levels and cardiovascular diseases (CVD) risk factors. CONCLUSIONS This study did not provide evidence that leptin levels are associated with increased risk of stroke and its subtypes.
Collapse
Affiliation(s)
- Qinqin Dai
- Nursing School of Zhengzhou University, Zhengzhou, Henan Province, China; The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yuanli Guo
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China; Nursing School of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Xiaofang Dong
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China; Nursing School of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yuan Gao
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China; Nursing School of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Shen Li
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Jiawei Zhao
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yuming Xu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China.
| | - Kai Liu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China; Nursing School of Zhengzhou University, Zhengzhou, Henan Province, China.
| |
Collapse
|
239
|
Xia M, Zhong Y, Peng Y, Qian C. Breakfast skipping and traits of cardiometabolic health: A mendelian randomization study. Clin Nutr ESPEN 2024; 59:328-333. [PMID: 38220394 DOI: 10.1016/j.clnesp.2023.12.149] [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: 07/19/2023] [Revised: 12/18/2023] [Accepted: 12/27/2023] [Indexed: 01/16/2024]
Abstract
BACKGROUND Breakfast skipping has been linked to poor cardiometabolic health in observational studies, but the causality remains unknown. Herein, we used Mendelian randomization (MR) approach to elucidate the potential causal effects of breakfast skipping on cardiometabolic traits. METHODS Genetic association estimates for breakfast skipping, cardiometabolic diseases, and cardiometabolic risk factors were extracted from the UK Biobank and several large genome-wide association studies. Two-sample MR analyses were performed primarily using the inverse variance weighted method, followed by sensitivity analysis to test the reliability of results. RESULTS MR results indicated no causal relationship between breakfast shipping with coronary heart disease (odds ratio [OR]: 1.079, 95 % confidence interval [CI]: 0.817-1.426; p = 0.591), stroke (OR: 0.877, 95 % CI: 0.680-1.131; p = 0.311), and type 2 diabetes mellitus (OR: 1.114, 95 % CI: 0.631-1.970; p = 0.709). However, genetically predicted breakfast skipping was significantly associated with increased body mass index (β: 0.250, standard error [SE]: 0.079; p = 0.001), waist-to-hip ratio (β: 0.177, SE: 0.076; p = 0.019), and low-density lipoprotein cholesterol (β: 0.260, SE: 0.115; p = 0.024). We found no evidence of association of genetic liability to breakfast skipping with blood pressure, glycemic traits, and other blood lipids. Sensitivity analysis supported the above results. CONCLUSION Our study suggested that breakfast skipping is causally linked to weight gain and higher serum low-density lipoprotein cholesterol, which may mediate the increased risk of cardiometabolic diseases reported in epidemiological studies.
Collapse
Affiliation(s)
- Meng Xia
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Yi Zhong
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Yongquan Peng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Cheng Qian
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China.
| |
Collapse
|
240
|
Alves M, Laranjeira F, Correia-da-Silva G. Understanding Hypertriglyceridemia: Integrating Genetic Insights. Genes (Basel) 2024; 15:190. [PMID: 38397180 PMCID: PMC10887881 DOI: 10.3390/genes15020190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/25/2024] Open
Abstract
Hypertriglyceridemia is an exceptionally complex metabolic disorder characterized by elevated plasma triglycerides associated with an increased risk of acute pancreatitis and cardiovascular diseases such as coronary artery disease. Its phenotype expression is widely heterogeneous and heavily influenced by conditions as obesity, alcohol consumption, or metabolic syndromes. Looking into the genetic underpinnings of hypertriglyceridemia, this review focuses on the genetic variants in LPL, APOA5, APOC2, GPIHBP1 and LMF1 triglyceride-regulating genes reportedly associated with abnormal genetic transcription and the translation of proteins participating in triglyceride-rich lipoprotein metabolism. Hypertriglyceridemia resulting from such genetic abnormalities can be categorized as monogenic or polygenic. Monogenic hypertriglyceridemia, also known as familial chylomicronemia syndrome, is caused by homozygous or compound heterozygous pathogenic variants in the five canonical genes. Polygenic hypertriglyceridemia, also known as multifactorial chylomicronemia syndrome in extreme cases of hypertriglyceridemia, is caused by heterozygous pathogenic genetic variants with variable penetrance affecting the canonical genes, and a set of common non-pathogenic genetic variants (polymorphisms, using the former nomenclature) with well-established association with elevated triglyceride levels. We further address recent progress in triglyceride-lowering treatments. Understanding the genetic basis of hypertriglyceridemia opens new translational opportunities in the scope of genetic screening and the development of novel therapies.
Collapse
Affiliation(s)
- Mara Alves
- Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal;
| | - Francisco Laranjeira
- CGM—Centro de Genética Médica Jacinto de Magalhães, Centro Hospitalar Universitário de Santo António (CHUdSA), 4099-028 Porto, Portugal;
- UMIB—Unit for Multidisciplinary Research in Biomedicine, ICBAS—School of Medicine and Biomedical Sciences, University of Porto, 4050-346 Porto, Portugal
- ITR—Laboratory for Integrative and Translational Research in Population Health, 4050-600 Porto, Portugal
| | - Georgina Correia-da-Silva
- UCIBIO Applied Molecular Biosciences Unit and Associate Laboratory i4HB—Institute for Health and Bioeconomy Laboratory of Biochemistry, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
241
|
Huang K, Huang S, Xiong M. Correlations between genetically predicted lipid-lowering drug targets and inflammatory bowel disease. Lipids Health Dis 2024; 23:31. [PMID: 38287401 PMCID: PMC10823737 DOI: 10.1186/s12944-024-02026-y] [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: 12/12/2023] [Accepted: 01/21/2024] [Indexed: 01/31/2024] Open
Abstract
BACKGROUND Millions of individuals globally suffer from Inflammatory bowel diseases (IBDs). There is a dearth of large population-based investigations on lipid metabolism and IBDs, and it is unclear whether lipid-lowering drugs target IBDs causally. Consequently, the aim of this study was to investigate the effects of lipid-lowering medication targets on the occurrence and progression of IBDs. METHODS Among the more than 400,000 participants in the UK Biobank cohort and the more than 170,000 participants in the Global Lipids Genetics Consortium, a total of nine genes linked to lipid-lowering drug targets were obtained (ABCG5/ABCG8, APOB, APOC3, LDLR, LPL, HMGCR, NPC1L1, PCSK9, and PPARA). IBD data were acquired from de Lange et al. (patients/sample size of IBDs: 25042/59957; ulcerative colitis (UC): 12366/45,975; Crohn's disease (CD): 12194/40,266) and the FinnGen cohort (patients/total sample size of IBDs: 4420/176,899; CD: 1520/171,906; UC: 3325/173,711). All four datasets were cross-combined for validation via Mendelian randomization analysis, and potential mediating factors were explored via mediation analysis. RESULTS Genetically proxied APOC3 inhibition was related to increased IBD risk (odds ratio (95% confidence interval): 0.87 (0.80-0.95); P < 0.01) and UC risk (0.83 (0.73-0.94); P < 0.01). IBD and CD risk were reduced by genetic mimicry of LDLR and LPL enhancements, respectively (odds ratioLDLR: 1.18 (1.03-1.36); P = 0.018; odds ratioCD: 1.26 (1.11-1.43); P = 2.60E-04). Genetically proxied HMGCR inhibition was associated with increased CD risk (0.68 (0.50-0.94); P = 0.018). These findings were confirmed through Mendelian analysis of the cross-combination of four separate datasets. APOC3-mediated triglyceride levels may contribute to IBDs partly through mediated triglycerides, Clostridium sensu stricto 1, Clostridiaceae 1, or the Lachnospiraceae FCS020 group. LDLR enhancement may contribute to IBDs partly through increasing Lactobacillaceae. CONCLUSION Vigilance is required to prevent adverse effects on IBDs (UC) for patients receiving volanesorsen (an antisense oligonucleotide targeting ApoC3 mRNA) and adverse effects on CD for statin users. LPL and LDLR show promise as candidate drug targets for CD and IBD, respectively, with mechanisms that are potentially independent of their lipid-lowering effects.
Collapse
Affiliation(s)
- Kuiyuan Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Jiangxi, 330000, China
| | - Shenan Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Jiangxi, 330000, China
| | - Ming Xiong
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Jiangxi, 330000, China.
| |
Collapse
|
242
|
Terasaki M, Izumi M, Yamagishi SI. A Clinical Case of Probable Sitosterolemia. Int J Mol Sci 2024; 25:1535. [PMID: 38338819 PMCID: PMC10855567 DOI: 10.3390/ijms25031535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Sitosterolemia is a rare genetic lipid disorder characterized by elevated plant sterols in the serum. A 24-year-old Japanese woman was referred to our hospital due to a high serum low-density lipoprotein cholesterol (LDL-C) level of 332 mg/dL. At first, she was suspected to suffer from familial hypercholesterolemia, and thus received lipid-lowering agents. Although her LDL-C level remained high (220 mg/dL) with diet therapy plus 10 mg/day rosuvastatin, it was drastically decreased to 46 mg/dL with the addition of 10 mg/day ezetimibe. Finally, her LDL-C level was well-controlled at about 70 mg/dL with 10 mg/day ezetimibe alone. Furthermore, while her serum sitosterol level was elevated at 10.5 μg/mL during the first visit to our hospital, it decreased to 3.6 μg/mL with the 10 mg/day ezetimibe treatment alone. These observations suggest that she might probably suffer from sitosterolemia. Therefore, targeted gene sequencing analysis was performed using custom panels focusing on the exome regions of 21 lipid-associated genes, including ABCG5, ABCG8, and familial hypercholesterolemia-causing genes (LDL receptor, LDLRAP1, PCSK9, and apolipoprotein B). We finally identified a heterozygous ABCG8 variant (NM_022437.2:c.1285A>G or NP_071882.1:p.Met429Val) in our patient. The same gene mutation was detected in her mother. We report here a rare case exhibiting probable sitosterolemia caused by a heterozygous Met429Val variant in the ABCG8 gene and additional unknown variants.
Collapse
Affiliation(s)
- Michishige Terasaki
- Division of Diabetes, Metabolism and Endocrinology, Showa University Graduate School of Medicine, 1-5-8 Shinagawa, Tokyo 142-8666, Japan;
| | - Mikiko Izumi
- Center for Clinical Genetics, Showa University Hospital, 1-5-8 Shinagawa, Tokyo 142-8666, Japan;
| | - Sho-ichi Yamagishi
- Division of Diabetes, Metabolism and Endocrinology, Showa University Graduate School of Medicine, 1-5-8 Shinagawa, Tokyo 142-8666, Japan;
| |
Collapse
|
243
|
Nebert DW. Gene-Environment Interactions: My Unique Journey. Annu Rev Pharmacol Toxicol 2024; 64:1-26. [PMID: 37788491 DOI: 10.1146/annurev-pharmtox-022323-082311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
I am deeply honored to be invited to write this scientific autobiography. As a physician-scientist, pediatrician, molecular biologist, and geneticist, I have authored/coauthored more than 600 publications in the fields of clinical medicine, biochemistry, biophysics, pharmacology, drug metabolism, toxicology, molecular biology, cancer, standardized gene nomenclature, developmental toxicology and teratogenesis, mouse genetics, human genetics, and evolutionary genomics. Looking back, I think my career can be divided into four distinct research areas, which I summarize mostly chronologically in this article: (a) discovery and characterization of the AHR/CYP1 axis, (b) pharmacogenomics and genetic prediction of response to drugs and other environmental toxicants, (c) standardized drug-metabolizing gene nomenclature based on evolutionary divergence, and (d) discovery and characterization of the SLC39A8 gene encoding the ZIP8 metal cation influx transporter. Collectively, all four topics embrace gene-environment interactions, hence the title of my autobiography.
Collapse
Affiliation(s)
- Daniel W Nebert
- Department of Environmental and Public Health Sciences and Center for Environmental Genetics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
- Department of Pediatrics and Molecular Developmental Biology, Division of Human Genetics, Cincinnati Children's Hospital, Cincinnati, Ohio, USA;
| |
Collapse
|
244
|
Hubacek JA, Adamkova V, Lanska V, Staněk V, Mrázková J, Gebauerová M, Kettner J, Kautzner J, Pitha J. Cholesterol associated genetic risk score and acute coronary syndrome in Czech males. Mol Biol Rep 2024; 51:164. [PMID: 38252350 PMCID: PMC10803395 DOI: 10.1007/s11033-023-09128-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 12/07/2023] [Indexed: 01/23/2024]
Abstract
BACKGROUND Despite a general decline in mean levels across populations, LDL-cholesterol levels remain a major risk factor for acute coronary syndrome (ACS). The APOB, LDL-R, CILP, and SORT-1 genes have been shown to contain variants that have significant effects on plasma cholesterol levels. METHODS AND RESULTS We examined polymorphisms within these genes in 1191 controls and 929 patients with ACS. Only rs646776 within SORT-1 was significantly associated with a risk of ACS (P < 0.05, AA vs. + G comparison; OR 1.21; 95% CI 1.01-1.45). With regard to genetic risk score (GRS), the presence of at least 7 alleles associated with elevated cholesterol levels was connected with increased risk (P < 0.01) of ACS (OR 1.26; 95% CI 1.06-1.52). Neither total mortality nor CVD mortality in ACS subjects (follow up-9.84 ± 3.82 years) was associated with the SNPs analysed or cholesterol-associated GRS. CONCLUSIONS We conclude that, based on only a few potent SNPs known to affect plasma cholesterol, GRS has the potential to predict ACS risk, but not ACS associated mortality.
Collapse
Affiliation(s)
- Jaroslav A Hubacek
- Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, IKEM-CEM-LMG, Videnska 1958/9, 140 21, Prague 4, Czech Republic.
- 3rd Department of Internal Medicine, 1st Faculty of Medicine, Charles University, Prague, Czech Republic.
| | - Vera Adamkova
- Preventive Cardiology Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Vera Lanska
- Information Technology Division, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Vladimir Staněk
- Cardiac Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Jolana Mrázková
- Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, IKEM-CEM-LMG, Videnska 1958/9, 140 21, Prague 4, Czech Republic
| | - Marie Gebauerová
- Cardiac Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Jiri Kettner
- Cardiac Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Josef Kautzner
- Cardiac Centre, Institute for Clinical and Experimental Medicine, Prague, Czech Republic
| | - Jan Pitha
- Experimental Medicine Centre, Institute for Clinical and Experimental Medicine, IKEM-CEM-LMG, Videnska 1958/9, 140 21, Prague 4, Czech Republic
| |
Collapse
|
245
|
Mahedy L, Anderson EL, Tilling K, Thornton ZA, Elmore AR, Szalma S, Simen A, Culp M, Zicha S, Harel BT, Davey Smith G, Smith EN, Paternoster L. Investigation of genetic determinants of cognitive change in later life. Transl Psychiatry 2024; 14:31. [PMID: 38238328 PMCID: PMC10796929 DOI: 10.1038/s41398-023-02726-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 12/14/2023] [Accepted: 12/22/2023] [Indexed: 01/22/2024] Open
Abstract
Cognitive decline is a major health concern and identification of genes that may serve as drug targets to slow decline is important to adequately support an aging population. Whilst genetic studies of cross-sectional cognition have been carried out, cognitive change is less well-understood. Here, using data from the TOMMORROW trial, we investigate genetic associations with cognitive change in a cognitively normal older cohort. We conducted a genome-wide association study of trajectories of repeated cognitive measures (using generalised estimating equation (GEE) modelling) and tested associations with polygenic risk scores (PRS) of potential risk factors. We identified two genetic variants associated with change in attention domain scores, rs534221751 (p = 1 × 10-8 with slope 1) and rs34743896 (p = 5 × 10-10 with slope 2), implicating NCAM2 and CRIPT/ATP6V1E2 genes, respectively. We also found evidence for the association between an education PRS and baseline cognition (at >65 years of age), particularly in the language domain. We demonstrate the feasibility of conducting GWAS of cognitive change using GEE modelling and our results suggest that there may be novel genetic associations for cognitive change that have not previously been associated with cross-sectional cognition. We also show the importance of the education PRS on cognition much later in life. These findings warrant further investigation and demonstrate the potential value of using trial data and trajectory modelling to identify genetic variants associated with cognitive change.
Collapse
Affiliation(s)
- Liam Mahedy
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, BS8 2BN, UK
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | - Emma L Anderson
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, BS8 2BN, UK
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | - Kate Tilling
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, BS8 2BN, UK
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston, NHS Foundation Trust and University of Bristol, Bristol, BS8 2BN, UK
| | - Zak A Thornton
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, BS8 2BN, UK
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
| | - Andrew R Elmore
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, BS8 2BN, UK
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston, NHS Foundation Trust and University of Bristol, Bristol, BS8 2BN, UK
| | - Sándor Szalma
- Takeda Development Center Americas, Inc., San Diego, CA, USA
| | - Arthur Simen
- Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Meredith Culp
- Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Stephen Zicha
- Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - Brian T Harel
- Takeda Development Center Americas, Inc., Cambridge, MA, USA
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, BS8 2BN, UK
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston, NHS Foundation Trust and University of Bristol, Bristol, BS8 2BN, UK
| | - Erin N Smith
- Takeda Development Center Americas, Inc., San Diego, CA, USA
| | - Lavinia Paternoster
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, BS8 2BN, UK.
- Department of Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS8 2BN, UK.
- NIHR Bristol Biomedical Research Centre, University Hospitals Bristol and Weston, NHS Foundation Trust and University of Bristol, Bristol, BS8 2BN, UK.
| |
Collapse
|
246
|
Chen Y, Kong W, Liu M, Li Q, Wang Y, Zheng Y, Zhou Y. Metabolic syndrome and risk of colorectal cancer: A Mendelian randomization study. Heliyon 2024; 10:e23872. [PMID: 38223733 PMCID: PMC10784169 DOI: 10.1016/j.heliyon.2023.e23872] [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: 04/01/2023] [Revised: 12/01/2023] [Accepted: 12/14/2023] [Indexed: 01/16/2024] Open
Abstract
Background Observational studies have previously demonstrated a significant relationship among both metabolic syndrome (Mets) and colorectal cancer (CRC). Whether there is a causal link remains controversial. Objective To clarify whether Mets and their components have a causal effect on colorectal cancer, we have carried out a bidirectional Mendelian randomization analysis (MR). Methods This study started from genome-wide association data for Mets and its 5 components (hypertension, waist circumference, fasting blood glucose, serum triglycerides, and serum high-density lipoprotein cholesterol) and colorectal cancer. Mendelian randomization (MR) techniques were used in the study to examine their associations. Results After Benjamini-Hochberg multiple corrections, genetically predicted significant causal link exists between WC (waist circumference) and CRC. The OR was 1.35 (95 % CI: 1.08-1.69; p = 0.0096). Other Mets components (HBP, FBG, TG, HDL), on the other hand, found no evidence of a genetic link between CRC and Mets. In addition, MR results showed that CRC was not causally related to either Mets or the components. We get the same result in the validated dataset. Conclusion According to the bidirectional MR investigation shows a significant causal relationship among obesity and CRC in the Mets component but no causal relationship in the opposite direction.
Collapse
Affiliation(s)
- Yuhua Chen
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Wanru Kong
- Department of Infection Management, Gansu Provincial Hospital, Lanzhou, 730000, China
| | - Min Liu
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Qiang Li
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yuping Wang
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Ya Zheng
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yongning Zhou
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory for Gastrointestinal Diseases of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
247
|
Zhao J, Chen R, Luo M, Gong H, Li K, Zhao Q. Lipid-lowering drugs and inflammatory bowel disease's risk: a drug-target Mendelian randomization study. Diabetol Metab Syndr 2024; 16:12. [PMID: 38191425 PMCID: PMC10775535 DOI: 10.1186/s13098-023-01252-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 12/31/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) has been associated with lipid-lowering drugs in observational studies. Drug-target Mendelian randomization (MR) was utilized in this study to examine the causal relationship between lipid-lowering drugs and incidence of IBD, aiming to identify new preventive uses for the drugs. METHODS We identified instrumental variables for three classes of lipid-lowering drugs: HMGCR inhibitors, PCSK9 inhibitors, and NPC1L1 inhibitors, using data from the Global Lipids Genetics Consortium. Summary statistics of IBD were obtained from UK Inflammatory Bowel Disease Genetics. The summary-data-based MR (SMR) and the inverse-variance weighted (IVW) MR were used for analysis. Sensitivity analyses were performed by conventional MR methods. RESULTS The SMR analysis showed no significant genetic association between increased gene expression of HMGCR, PCSK9, and NPC1L1 and IBD, Crohn's disease (CD) and ulcerative colitis (UC). According to IVW-MR analysis, increased HMGCR expression is associated with a reduced risk of IBD (OR = 0.73, 95% confidence interval (CI) 0.59-0.90, P = 0.003) and CD (OR = 0.75, 95% CI 0.57-0.97, P = 0.03), but not with UC. Additionally, increased NPC1L1 gene expression was associated with elevated risk of IBD (OR = 1.60, 95% CI 1.07-2.40, P = 0.023), but not with CD and UC. However, no significant causal relationships were found between PCSK9 gene expression and IBD, CD, and UC. The sensitivity analysis demonstrated no evidence of heterogeneity or pleiotropy among the reported results. CONCLUSIONS The heightened expression of genetic variations in HMGCR inhibitor targets could potentially reduce the risk of IBD and CD, while genetic variation in the expression of NPC1L1 targets was positively associated with IBD.
Collapse
Affiliation(s)
- Jiaxi Zhao
- General Practice Ward/International Medical Center Ward General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Rong Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Mengqi Luo
- Department of Gastroenterology and Hepatology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hongping Gong
- General Practice Ward/International Medical Center Ward General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Kaixin Li
- Department of Nephrology, Huadong Hospital, Shanghai, China
| | - Qian Zhao
- General Practice Ward/International Medical Center Ward General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
248
|
Peters U, Tomlinson I. Utilizing Human Genetics to Develop Chemoprevention for Cancer-Too Good an Opportunity to be Missed. Cancer Prev Res (Phila) 2024; 17:7-12. [PMID: 38173394 DOI: 10.1158/1940-6207.capr-22-0523] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 03/20/2023] [Accepted: 12/06/2023] [Indexed: 01/05/2024]
Abstract
Large-scale genetic studies are reliably identifying many risk factors for disease in the general population. Several of these genetic risk factors encode potential drug targets, and genetics has already helped to introduce targeted agents for some diseases, an example being lipid-lowering drugs to reduce the incidence of cardiovascular disease. Multiple drugs have been developed to treat cancers based on somatic mutations and genomics, but in stark contrast, there seems to be a reluctance to use germline genetic data to develop drugs to prevent malignancy, despite the large numbers of people who could benefit, the potential for lowering cancer rates, and the widespread current use of non-pharmaceutical measures to reduce cancer risk factors such as tobacco, alcohol, and infectious diseases. We argue that concerted efforts for cancer prevention based on genetics, including genes influenced by common polymorphisms that modulate cancer risk, are urgently needed. There are enormous, yet underutilized, opportunities to develop novel targeted agents for chemoprevention of cancer based on human germline genetics. Such efforts are likely to require the support of a dedicated funding program by national and international agencies. See related commentary by Winham and Sherman, p. 13.
Collapse
Affiliation(s)
- Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Center and Department of Epidemiology, University of Washington, Seattle, Washington
| | - Ian Tomlinson
- Department of Oncology, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
249
|
Grant AJ, Burgess S. A Bayesian approach to Mendelian randomization using summary statistics in the univariable and multivariable settings with correlated pleiotropy. Am J Hum Genet 2024; 111:165-180. [PMID: 38181732 PMCID: PMC10806746 DOI: 10.1016/j.ajhg.2023.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 12/01/2023] [Accepted: 12/01/2023] [Indexed: 01/07/2024] Open
Abstract
Mendelian randomization uses genetic variants as instrumental variables to make causal inferences on the effect of an exposure on an outcome. Due to the recent abundance of high-powered genome-wide association studies, many putative causal exposures of interest have large numbers of independent genetic variants with which they associate, each representing a potential instrument for use in a Mendelian randomization analysis. Such polygenic analyses increase the power of the study design to detect causal effects; however, they also increase the potential for bias due to instrument invalidity. Recent attention has been given to dealing with bias caused by correlated pleiotropy, which results from violation of the "instrument strength independent of direct effect" assumption. Although methods have been proposed that can account for this bias, a number of restrictive conditions remain in many commonly used techniques. In this paper, we propose a Bayesian framework for Mendelian randomization that provides valid causal inference under very general settings. We propose the methods MR-Horse and MVMR-Horse, which can be performed without access to individual-level data, using only summary statistics of the type commonly published by genome-wide association studies, and can account for both correlated and uncorrelated pleiotropy. In simulation studies, we show that the approach retains type I error rates below nominal levels even in high-pleiotropy scenarios. We demonstrate the proposed approaches in applied examples in both univariable and multivariable settings, some with very weak instruments.
Collapse
Affiliation(s)
- Andrew J Grant
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK; Sydney School of Public Health, University of Sydney, Sydney, NSW, Australia.
| | - Stephen Burgess
- MRC Biostatistics Unit, University of Cambridge, Cambridge, UK; Cardiovascular Epidemiology Unit, University of Cambridge, Cambridge, UK
| |
Collapse
|
250
|
Riesmeijer SA, Kamali Z, Ng M, Drichel D, Piersma B, Becker K, Layton TB, Nanchahal J, Nothnagel M, Vaez A, Hennies HC, Werker PMN, Furniss D, Nolte IM. A genome-wide association meta-analysis implicates Hedgehog and Notch signaling in Dupuytren's disease. Nat Commun 2024; 15:199. [PMID: 38172110 PMCID: PMC10764787 DOI: 10.1038/s41467-023-44451-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 12/13/2023] [Indexed: 01/05/2024] Open
Abstract
Dupuytren's disease (DD) is a highly heritable fibrotic disorder of the hand with incompletely understood etiology. A number of genetic loci, including Wnt signaling members, have been previously identified. Our overall aim was to identify novel genetic loci, to prioritize genes within the loci for functional studies, and to assess genetic correlation with associated disorders. We performed a meta-analysis of six DD genome-wide association studies from three European countries and extensive bioinformatic follow-up analyses. Leveraging 11,320 cases and 47,023 controls, we identified 85 genome-wide significant single nucleotide polymorphisms in 56 loci, of which 11 were novel, explaining 13.3-38.1% of disease variance. Gene prioritization implicated the Hedgehog and Notch signaling pathways. We also identified a significant genetic correlation with frozen shoulder. The pathways identified highlight the potential for new therapeutic targets and provide a basis for additional mechanistic studies for a common disorder that can severely impact hand function.
Collapse
Affiliation(s)
- Sophie A Riesmeijer
- University of Groningen, University Medical Center Groningen, Department of Plastic Surgery, Groningen, The Netherlands.
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, The Netherlands.
| | - Zoha Kamali
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, The Netherlands
- Department of bioinformatics, School of Advanced Medical Technologies, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Michael Ng
- Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Science, Botnar Research Centre, University of Oxford, Oxford, UK
| | - Dmitriy Drichel
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
- Faculty of Medicine and the Cologne University Hospital, Cologne, Germany
| | - Bram Piersma
- University of Groningen, Groningen, The Netherlands
| | - Kerstin Becker
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
| | | | | | - Michael Nothnagel
- Cologne Center for Genomics, University of Cologne, Cologne, Germany
- Faculty of Medicine and the Cologne University Hospital, Cologne, Germany
| | - Ahmad Vaez
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, The Netherlands
- Department of bioinformatics, School of Advanced Medical Technologies, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hans Christian Hennies
- Faculty of Medicine and the Cologne University Hospital, Cologne, Germany
- Department of Biological Sciences, University of Huddersfield, Huddersfield, UK
| | - Paul M N Werker
- University of Groningen, University Medical Center Groningen, Department of Plastic Surgery, Groningen, The Netherlands
| | - Dominic Furniss
- Nuffield Department of Orthopaedics, Rheumatology, and Musculoskeletal Science, Botnar Research Centre, University of Oxford, Oxford, UK
| | - Ilja M Nolte
- University of Groningen, University Medical Center Groningen, Department of Epidemiology, Groningen, The Netherlands
| |
Collapse
|