1
|
Shriner D, Bentley AR, Doumatey AP, Zhou J, Chen G, Rotimi CN, Adeyemo AA. Three Loci Affecting Variance of Body Mass Index in African Americans and Sub-Saharan Africans. Genet Epidemiol 2025; 49:e70009. [PMID: 40323147 PMCID: PMC12051743 DOI: 10.1002/gepi.70009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 01/08/2025] [Accepted: 04/16/2025] [Indexed: 05/07/2025]
Abstract
Conventional genome-wide association studies (GWAS) are designed to assess the effect of a genetic locus on phenotypic mean by genotype. Such loci explain a proportion of phenotypic variance known as narrow-sense heritability. In contrast, variance quantitative trait loci (vQTL) are associated with the phenotypic variance by genotype. These loci explain an additional proportion of phenotypic variance and contribute to broad-sense heritability but not to narrow-sense heritability. Here, a genome-wide vQTL analysis in 22,805 African Americans yielded eight loci for body mass index (BMI). Of these loci, three were replicated in 6002 sub-Saharan Africans. No locus reached genome-wide significance using the standard additive model. Furthermore, no locus showed evidence for natural selection, haplotype effects, or gene × sex or gene × study interactions. Two loci showed evidence for an effect of locus-specific ancestry resulting from admixture and for a gene × gene interaction. One locus showed evidence for interaction with diastolic blood pressure, consistent with this vQTL capturing an unmodeled gene × covariate interaction. These analyses demonstrate that relevant BMI loci can be detected by evaluating vQTL and that these loci contribute to the underexplored broad-sense heritability for this trait.
Collapse
Affiliation(s)
- Daniel Shriner
- Center for Research on Genomics and Global HealthNational Human Genome Research InstituteBethesdaMarylandUSA
| | - Amy R. Bentley
- Center for Research on Genomics and Global HealthNational Human Genome Research InstituteBethesdaMarylandUSA
| | - Ayo P. Doumatey
- Center for Research on Genomics and Global HealthNational Human Genome Research InstituteBethesdaMarylandUSA
| | - Jie Zhou
- Center for Research on Genomics and Global HealthNational Human Genome Research InstituteBethesdaMarylandUSA
| | - Guanjie Chen
- Center for Research on Genomics and Global HealthNational Human Genome Research InstituteBethesdaMarylandUSA
| | - Charles N. Rotimi
- Center for Research on Genomics and Global HealthNational Human Genome Research InstituteBethesdaMarylandUSA
| | - Adebowale A. Adeyemo
- Center for Research on Genomics and Global HealthNational Human Genome Research InstituteBethesdaMarylandUSA
| |
Collapse
|
2
|
Lee H, Fernandes M, Lee J, Merino J, Kwak SH. Exploring the shared genetic landscape of diabetes and cardiovascular disease: findings and future implications. Diabetologia 2025; 68:1087-1100. [PMID: 40088285 PMCID: PMC12069157 DOI: 10.1007/s00125-025-06403-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 01/28/2025] [Indexed: 03/17/2025]
Abstract
Diabetes is a rapidly growing global health concern projected to affect one in eight adults by 2045, which translates to roughly 783 million people. The profound metabolic alterations often present in dysglycaemia significantly increase the risk of cardiovascular complications. While genetic susceptibility plays a crucial role in diabetes and its vascular complications, identifying genes and molecular mechanisms that influence both diseases simultaneously has proven challenging. A key reason for this challenge is the pathophysiological heterogeneity underlying these diseases, with multiple processes contributing to different forms of diabetes and specific cardiovascular complications. This molecular heterogeneity has limited the effectiveness of large-scale genome-wide association studies (GWAS) in identifying shared underlying mechanisms. Additionally, our limited knowledge of the causal genes, cell types and disease-relevant states through which GWAS signals operate has hindered the discovery of common molecular pathways. This review highlights recent advances in genetic epidemiology, including studies of causal associations that have uncovered genetic and molecular factors influencing both dysglycaemia and cardiovascular complications. We explore how disease subtyping approaches can be critical in pinpointing the unique molecular signatures underlying both diabetes and cardiovascular complications. Finally, we address critical research gaps and future opportunities to advance our understanding of both diseases and translate these discoveries into tangible benefits for patient care and population health.
Collapse
Affiliation(s)
- Hyunsuk Lee
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Hospital, Seoul, Korea
- Department of Translational Medicine, Seoul National University College of Medicine, Seoul, Korea
- Genomic Medicine Institute, Medical Research Center, Seoul National University College of Medicine, Seoul, Korea
| | - Maria Fernandes
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Jeongeun Lee
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Hospital, Seoul, Korea
| | - Jordi Merino
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.
- Diabetes Unit, Endocrine Division, Massachusetts General Hospital, Boston, MA, USA.
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
| | - Soo Heon Kwak
- Department of Internal Medicine, Seoul National University College of Medicine and Seoul National University Hospital, Seoul, Korea.
| |
Collapse
|
3
|
Lin M, Guo J, Tao H, Gu Z, Tang W, Zhou F, Jiang Y, Zhang R, Jia D, Sun Y, Jia P. Circulating mediators linking cardiometabolic diseases to HFpEF: a mediation Mendelian randomization analysis. Cardiovasc Diabetol 2025; 24:201. [PMID: 40355922 PMCID: PMC12070650 DOI: 10.1186/s12933-025-02738-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 04/10/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) is an increasingly prevalent clinical syndrome with high morbidity and mortality. Although HFpEF frequently coexists with cardiometabolic diseases, the causal mechanisms and potential mediators remain poorly understood. OBJECTIVES This study aimed to identify cardiometabolic risk factors specifically driving HFpEF and to determine their underlying circulating mediators. METHODS We used two-sample Mendelian Randomization (MR) to analyze the effects of obesity, Type 2 diabetes, hypertension, chronic kidney disease (CKD), and dyslipidemia on HFpEF and heart failure with reduced ejection fraction (HFrEF) in large European-ancestry GWAS datasets. We then performed mediation MR to identify plasma proteins and metabolites that mediate the transition from each cardiometabolic disease to HFpEF, respectively. We applied multivariable MR to assess the impact of risk confounding on the results. Bioinformatic analyses were conducted to delineate mechanisms. RESULTS Cardiometabolic diseases had heterogeneous effects on HFpEF and HFrEF. Obesity and type 2 diabetes showed adjusted causal effects with HFpEF, hypertension showed potential relevance to HFpEF, whereas dyslipidemia and CKD did not. MR analysis identified 5 proteins that mediate obesity to HFpEF; 5 proteins that mediate type 2 diabetes to HFpEF. Further mediation MR analysis of obesity and T2D on HFrEF revealed heterogeneity in circulating mediators between metabolic HFpEF and HFrEF. Comprehensive bioinformatics analyses showed that IL1R1, together with other proteins such as TP53 and FGF19, orchestrates the inflammatory and fibrotic processes underlying HFpEF. CONCLUSIONS These findings suggest that metabolic HFpEF has distinct etiological features compared with HFrEF and is driven by complex, condition-specific mediators. IL1R1 mediates HFpEF in multiple metabolic risk states, suggesting a potential therapeutic target. Further translational studies are warranted to evaluate anti-inflammatory strategies targeting IL1R1 in HFpEF.
Collapse
MESH Headings
- Humans
- Mendelian Randomization Analysis
- Heart Failure/genetics
- Heart Failure/physiopathology
- Heart Failure/blood
- Heart Failure/epidemiology
- Heart Failure/diagnosis
- Cardiometabolic Risk Factors
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/epidemiology
- Diabetes Mellitus, Type 2/diagnosis
- Risk Assessment
- Stroke Volume/genetics
- Biomarkers/blood
- Obesity/genetics
- Obesity/blood
- Obesity/epidemiology
- Obesity/diagnosis
- Dyslipidemias/genetics
- Dyslipidemias/blood
- Dyslipidemias/epidemiology
- Dyslipidemias/diagnosis
- Hypertension/genetics
- Hypertension/blood
- Hypertension/epidemiology
- Hypertension/diagnosis
- Mediation Analysis
- Ventricular Function, Left/genetics
- Renal Insufficiency, Chronic/genetics
- Renal Insufficiency, Chronic/blood
- Renal Insufficiency, Chronic/epidemiology
- Renal Insufficiency, Chronic/diagnosis
- Genetic Predisposition to Disease
- Phenotype
- Genome-Wide Association Study
- Databases, Genetic
Collapse
Affiliation(s)
- Mingzhi Lin
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Jiuqi Guo
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Hongqian Tao
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Zhilin Gu
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Wenyi Tang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Fuliang Zhou
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Yanling Jiang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Ruyi Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Dalin Jia
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China.
| | - Yingxian Sun
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China.
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang, Liaoning, China.
| | - Pengyu Jia
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China.
| |
Collapse
|
4
|
Chen S, Liang Y, Mo JMY, Li QHY, He B, Luo S, Burgess S, Au Yeung SL. Challenges in interpreting Mendelian randomization studies with a disease as the exposure: Using COVID-19 liability studies as an exemplar. Eur J Hum Genet 2025; 33:658-665. [PMID: 40164729 PMCID: PMC12048694 DOI: 10.1038/s41431-025-01840-x] [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: 07/20/2024] [Revised: 02/14/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025] Open
Abstract
Mendelian randomization (MR) studies using diseases as exposures are increasingly prevalent although any observed associations do not necessarily imply effect of diseases. To illustrate this challenge, we conducted a systematic review of MR studies focusing on COVID-19 consequence. We hypothesized if outcome genome-wide association studies (GWAS) were conducted before COVID-19 pandemic in late 2019, any observed associations in these studies were unlikely to be driven by COVID-19. We systematically searched PubMed, EMBASE, and MEDLINE for all MR studies published between 1 January 2019 and 20 May 2023. Inclusion criteria included MR studies which used COVID-19 as the exposure and designed to assess COVID-19's impact on health outcomes. We extracted relevant information, such as result interpretation and relevance assumption assessment. This review was registered at PROSPERO (CRD42023421079). Amongst 57 included studies, 45 studies used outcome GWAS published prior to 2019 whilst the remaining studies likely used outcome GWAS containing data collected before 2019. Relevance assumption was assessed mainly by p values. A total of 35 studies showed an association of COVID-19 liability with health outcomes. Regardless of the results, 45 studies attributed these as evidence (or lack of evidence) of COVID-19 consequence. In MR studies using disease liability as exposure, relevance assumption should consider the prevalence of the disease in the outcome GWAS in the context of 2 sample Mendelian randomization study rather than p values/F-statistic alone. Even when these are verified, these studies likely suffered from pleiotropy, making corresponding interpretation as effect of disease challenging.
Collapse
Affiliation(s)
- Siyu Chen
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
| | - Ying Liang
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
| | - Jacky Man Yuen Mo
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
| | - Queenie Ho Yi Li
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
| | - Baoting He
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
| | - Shan Luo
- Department of Family Medicine and Primary Care, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China
| | - Stephen Burgess
- MRC Biostatistics Unit, Cambridge Institute of Public Health, University of Cambridge, Cambridge, United Kingdom
| | - Shiu Lun Au Yeung
- School of Public Health, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong Special Administrative Region, Hong Kong, China.
| |
Collapse
|
5
|
Cao T, Zhou Q, Li F, Wang M, Zhang M, Li X, Zhao H, Zhou Y. Dual-specific phosphatases-8: a new target for clinical disease intervention. J Transl Med 2025; 23:485. [PMID: 40301852 PMCID: PMC12042392 DOI: 10.1186/s12967-025-06499-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: 01/21/2025] [Accepted: 04/13/2025] [Indexed: 05/01/2025] Open
Abstract
Dual-specific phosphatase-8 (DUSP8), identified as the first gene in a genome-wide association study (GWAS), is implicated in cellular oxidative stress, proliferation, apoptosis, and drug resistance through its negative regulation of the dephosphorylation activities of JNK, ERK, and p38 within the MAPK pathway. Recent studies have shown that DUSP8 plays a pivotal role in the progression of several human diseases, notably colorectal cancer, diabetic kidney disease, and breast cancer. This suggests that DUSP8 may represent a novel target for clinical intervention in these diseases. This review first introduces the biological structure and function of DUSP8, with a focus on its relationship with a series of diseases and the regulatory mechanisms involved. Furthermore, we concentrate on unresolved scientific questions in the current research, aiming to establish a new theoretical foundation for the diagnosis and treatment of related diseases.
Collapse
Affiliation(s)
- Tingping Cao
- Department of Pathophysiology, Zunyi Medical University, Zunyi, Guizhou, 563000, China
- Department of Physics, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Quanling Zhou
- Department of Pathophysiology, Zunyi Medical University, Zunyi, Guizhou, 563000, China
- Department of Physics, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Fujun Li
- Department of Pathophysiology, Zunyi Medical University, Zunyi, Guizhou, 563000, China
- Department of Physics, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Mingyue Wang
- Department of Pathophysiology, Zunyi Medical University, Zunyi, Guizhou, 563000, China
- Department of Physics, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Ming Zhang
- Department of Physics, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Xiaohui Li
- Department of Physics, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Hailong Zhao
- Department of Pathophysiology, Zunyi Medical University, Zunyi, Guizhou, 563000, China
| | - Ya Zhou
- Department of Pathophysiology, Zunyi Medical University, Zunyi, Guizhou, 563000, China.
- Department of Physics, Zunyi Medical University, Zunyi, Guizhou, 563000, China.
- Key Laboratory of Cancer Prevention and Treatment of Guizhou Province, Zunyi, Guizhou, 563000, China.
| |
Collapse
|
6
|
de Vries P, Hasbani N, Heath A, Hodonsky C, Hahn J, Meena D, Lu H, Dehghan AA, Kavousi M, Voight B, Peyser P, Morrison A, Assimes T, Damrauer S, Miller C. A multi-trait genome-wide association study of coronary artery disease and subclinical atherosclerosis traits. RESEARCH SQUARE 2025:rs.3.rs-6456056. [PMID: 40313769 PMCID: PMC12045367 DOI: 10.21203/rs.3.rs-6456056/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Measures of subclinical atherosclerosis, such as coronary artery calcification (CAC) and carotid intima-media thickness (CIMT), reflect the underlying pathophysiology of coronary artery disease (CAD) and are genetically correlated with CAD and related risk factors. Leveraging summary statistics from genome-wide association studies of CAD, CIMT, CAC, type 2 diabetes, low-density lipoprotein cholesterol, and systolic blood pressure, we performed 15 separate multi-trait GWAS to identify shared susceptibility loci and elucidate the pleiotropic architecture underlying atherosclerosis. We identified 442 shared risk loci across all analyses that met an experiment-wide Bonferroni threshold of 3.3 × 10-9, uncovering 195 novel atherosclerosis loci. Multi-trait colocalization confirmed a shared causal signal in 25 shared novel loci for atherosclerosis. Trait-eQTL colocalization identified evidence of a shared causal signal in arterial, subcutaneous adipose, and cardiac tissues, implicating genes such as PRRX2, BNC2, CLIC4, SCAI, and PPP6C, and pathways related to vascular remodeling, inflammation, and metabolic regulation.
Collapse
Affiliation(s)
| | | | - Adam Heath
- The University of Texas Health Science Center at Houston
| | | | - Julie Hahn
- The University of Texas Health Science Center at Houston
| | | | | | | | | | | | - Patricia Peyser
- Department of Epidemiology, School of Public Health, University of Michigan
| | | | | | | | | |
Collapse
|
7
|
Gofron KK, Wasilewski A, Małgorzewicz S. Effects of GLP-1 Analogues and Agonists on the Gut Microbiota: A Systematic Review. Nutrients 2025; 17:1303. [PMID: 40284168 PMCID: PMC12029897 DOI: 10.3390/nu17081303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND GLP-1 analogues are a relatively new class of medications that form the cornerstone of diabetes treatment. They possess invaluable glucose-lowering properties without hypoglycemic effects as well as strong cardioprotective effects. The gut microbiome has become the focus of numerous studies, demonstrating its influence not only on the gut but also on the overall well-being of the entire body. However, the effects of GLP-1 analogs on gut microbiota remain uncertain. SCOPE OF REVIEW Our systematic review (based on PRISMA guidelines) aimed to gather knowledge on the effects of GLP-1 analogue medications on the composition, richness, and abundance of gut microbiota in both animal and human models. CONCLUSIONS Thirty-eight studies were included in this systematic review. GLP-1 analogues have demonstrated a notable impact on the composition, richness, and diversity of gut microbiota. We can conclude, following the obtained research results of our study, that liraglutide promotes the growth of beneficial genera relevant for beneficial metabolic functions. Exenatide and exendin-4 administration showed various effects on the microbiome composition in animal and human studies. In animal models, it increased genera associated with improved metabolism; however, in human models, genera linked to better metabolic functions and escalated inflammation increased. Following dulaglutide administration, increases in Bacteroides, Akkermansia, and Ruminococcus, genera connected to an improved metabolic model, were significant. Finally, varied results were obtained after semaglutide treatment, in which A. muciniphila, known for its positive metabolic functions, increased; however, microbial diversity decreased. Semaglutide treatment provided various results indicating many confounding factors in semaglutide's impact on the gut microbiota. Results varied due to dissimilarities in the studied populations and the duration of the studies. Further research is essential to confirm these findings and to better recognize their implications for the clinical outcomes of patients.
Collapse
Affiliation(s)
- Krzysztof Ksawery Gofron
- Student Scientific Circle at Department of Clinical Nutrition, Medical University of Gdańsk, Marii Skłodowskiej-Curie 3a, 80-210 Gdańsk, Poland
| | - Andrzej Wasilewski
- Student Scientific Association of Medical Chemistry and Immunochemistry, Wroclaw Medical University, Marii Skłodowskiej-Curie 48/50, 59-369 Wroclaw, Poland;
| | - Sylwia Małgorzewicz
- Department of Clinical Nutrition, Medical University of Gdańsk, Marii Skłodowskiej-Curie 3a, 80-210 Gdańsk, Poland;
- Department of Nephrology, Transplantology, and Internal Medicine, Medical University of Gdańsk, Marii Skłodowskiej-Curie 3a, 80-210 Gdańsk, Poland
| |
Collapse
|
8
|
Tang B, Lin N, Liang J, Yi G, Zhang L, Peng W, Xue C, Jiang H, Li M. Leveraging pleiotropic clustering to address high proportion correlated horizontal pleiotropy in Mendelian randomization studies. Nat Commun 2025; 16:2817. [PMID: 40118820 PMCID: PMC11928562 DOI: 10.1038/s41467-025-57912-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 03/05/2025] [Indexed: 03/24/2025] Open
Abstract
Mendelian randomization harnesses genetic variants as instrumental variables to infer causal relationships between exposures and outcomes. However, certain genetic variants can affect both the exposure and the outcome through a shared factor. This phenomenon, called correlated horizontal pleiotropy, may result in false-positive causal findings. Here, we propose a Pleiotropic Clustering framework for Mendelian randomization, PCMR. PCMR detects correlated horizontal pleiotropy and extends the zero modal pleiotropy assumption to enhance causal inference in trait pairs with correlated horizontal pleiotropic variants. Simulations show that PCMR can effectively detect correlated horizontal pleiotropy and avoid false positives in the presence of correlated horizontal pleiotropic variants, even when they constitute a high proportion of the variants connecting both traits (e.g., 30-40%). In datasets consisting of 48 exposure-common disease pairs, PCMR detects horizontal correlated pleiotropy in 7 out of the exposure-common disease pairs, and avoids detecting false positive causal links. Additionally, PCMR can facilitate the integration of biological information to exclude correlated horizontal pleiotropic variants, enhancing causal inference. We apply PCMR to study causal relationships between three common psychiatric disorders as examples.
Collapse
Affiliation(s)
- Bin Tang
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Nan Lin
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Junhao Liang
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Guorong Yi
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Liubin Zhang
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Wenjie Peng
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Chao Xue
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Hui Jiang
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Miaoxin Li
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China.
- Center for Precision Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.
| |
Collapse
|
9
|
Wang M, Collings PJ, Day FR, Ong KK, Brage S, Sharp SJ, Jang H, Suh S, Luo S, Au Yeung SL, Kim Y. Genetic Susceptibility to Type 2 Diabetes, Television Viewing, and Atherosclerotic Cardiovascular Disease Risk. J Am Heart Assoc 2025; 14:e036811. [PMID: 40071666 DOI: 10.1161/jaha.124.036811] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 01/06/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND Type 2 diabetes (T2D) is a major risk factor for atherosclerotic cardiovascular disease (ASCVD). This study examined the interplay between watching television and T2D genetic risk for risk of ASCVD. METHODS We included 346 916 White British individuals from UK Biobank. A weighted polygenic risk score for T2D was calculated on the basis of 138 genetic variants associated with T2D. Time spent watching television was self-reported and categorized into 2 groups: ≤1 h/d and ≥2 h/d. Over a median 13.8-year follow-up, 21 265 incident ASCVD events were identified. Models using Cox regression with age as the underlying time scale adjusted for potential confounders (demographic, anthropometric, lifestyle factors, and medication use) were fit. RESULTS Compared with watching television for ≤1 h/d, watching ≥2 h/d was associated with 12% (95% CI, 1.07-1.16) higher hazards of ASCVD, independently of T2D genetic risk. Joint analyses (with low T2D genetic risk and ≤1 h/d of television viewing as reference) indicated that medium and high T2D genetic risk was not associated with higher hazards of ASCVD as long as television viewing was ≤1 h/d. The P values for multiplicative and additive interactions between T2D genetic risk and television viewing were 0.050 and 0.038, respectively. The 10-year absolute risk of ASCVD was lower for high T2D genetic risk combined with ≤1 h/d of television viewing (2.13%) than for low T2D genetic risk combined with ≥2 h/d of television viewing (2.46%). CONCLUSIONS Future clinical trials of lifestyle-modification interventions targeting specific types of screen-based sedentary activities could be implemented to individuals at high genetic risk of T2D for primary prevention of ASCVD.
Collapse
Affiliation(s)
- Mengyao Wang
- School of Public Health The University of Hong Kong Li Ka Shing Faculty of Medicine Hong Kong SAR China
| | - Paul James Collings
- School of Public Health The University of Hong Kong Li Ka Shing Faculty of Medicine Hong Kong SAR China
| | - Felix R Day
- MRC Epidemiology Unit Institute of Metabolic Science, University of Cambridge Cambridge Cambridgeshire United Kingdom
| | - Ken K Ong
- MRC Epidemiology Unit Institute of Metabolic Science, University of Cambridge Cambridge Cambridgeshire United Kingdom
| | - Soren Brage
- MRC Epidemiology Unit Institute of Metabolic Science, University of Cambridge Cambridge Cambridgeshire United Kingdom
| | - Stephen J Sharp
- MRC Epidemiology Unit Institute of Metabolic Science, University of Cambridge Cambridge Cambridgeshire United Kingdom
| | - Haeyoon Jang
- School of Public Health The University of Hong Kong Li Ka Shing Faculty of Medicine Hong Kong SAR China
| | - Siyeon Suh
- School of Public Health The University of Hong Kong Li Ka Shing Faculty of Medicine Hong Kong SAR China
| | - Shan Luo
- School of Public Health The University of Hong Kong Li Ka Shing Faculty of Medicine Hong Kong SAR China
| | - Shiu Lun Au Yeung
- School of Public Health The University of Hong Kong Li Ka Shing Faculty of Medicine Hong Kong SAR China
| | - Youngwon Kim
- School of Public Health The University of Hong Kong Li Ka Shing Faculty of Medicine Hong Kong SAR China
- MRC Epidemiology Unit Institute of Metabolic Science, University of Cambridge Cambridge Cambridgeshire United Kingdom
| |
Collapse
|
10
|
Chen HH, Highland HM, Frankel EG, Scartozzi AC, Zhang X, Roshani R, Sharma P, Kar A, Buchanan VL, Polikowsky HG, Petty LE, Seo J, Anwar MY, Kim D, Graff M, Young KL, Zhu W, Karastergiou K, Shaw DM, Justice AE, Fernández-Rhodes L, Krishnan M, Gutierrez A, McCormick PJ, Aguilar-Salinas CA, Tusié-Luna MT, Muñoz-Hernandez LL, Herrera-Hernandez M, Lee M, Gamazon ER, Cox NJ, Pajukanta P, Fried SK, Gordon-Larsen P, Shah RV, Fisher-Hoch SP, McCormick JB, North KE, Below JE. Multiomics reveal key inflammatory drivers of severe obesity: IL4R, LILRA5, and OSM. CELL GENOMICS 2025; 5:100784. [PMID: 40043711 PMCID: PMC11960538 DOI: 10.1016/j.xgen.2025.100784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 10/08/2024] [Accepted: 02/06/2025] [Indexed: 03/15/2025]
Abstract
Polygenic severe obesity (body mass index [BMI] ≥40 kg/m2) has increased, especially in Hispanic/Latino populations, yet we know little about the underlying mechanistic pathways. We analyzed whole-blood multiomics data to identify genes differentially regulated in severe obesity in Mexican Americans from the Cameron County Hispanic Cohort. Our RNA sequencing analysis identified 124 genes significantly differentially expressed between severe obesity cases (BMI ≥40 kg/m2) and controls (BMI <25 kg/m2); 33% replicated in an independent sample from the same population. Our integrative approach identified inflammatory genes, including IL4R, ZNF438, and LILRA5. Several genes displayed transcriptomic effects on severe obesity in subcutaneous adipose tissue. We further showed that the genetic regulation of these genes is associated with several traits in a large biobank, including bone fractures, obstructive sleep apnea, and hyperaldosteronism, illuminating potential risk mechanisms. Our findings furnish a molecular architecture of the severe obesity phenotype across multiple molecular domains.
Collapse
Affiliation(s)
- Hung-Hsin Chen
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; Academia Sinica, Institute of Biomedical Sciences, Taipei, Taiwan
| | - Heather M Highland
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Elizabeth G Frankel
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Alyssa C Scartozzi
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Xinruo Zhang
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Rashedeh Roshani
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Priya Sharma
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Asha Kar
- Department of Human Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, USA; Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, USA
| | - Victoria L Buchanan
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hannah G Polikowsky
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Lauren E Petty
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jungkyun Seo
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of MetaBiohealth, Sungkyunkwan University, Suwon, Republic of Korea
| | - Mohammad Yaser Anwar
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Daeeun Kim
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mariaelisa Graff
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kristin L Young
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Wanying Zhu
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Kalypso Karastergiou
- Obesity Research Center, Boston University School of Medicine, Boston, MA, USA; Diabetes Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Douglas M Shaw
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Anne E Justice
- Department of Population Health Services, Geisinger Health, Danville, PA, USA
| | | | - Mohanraj Krishnan
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Absalon Gutierrez
- Department of Internal Medicine, Division of Endocrinology, Diabetes, and Metabolism, Houston, TX, USA
| | - Peter J McCormick
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine, Queen Mary University of London, London, UK
| | - Carlos A Aguilar-Salinas
- Unidad de Investigación de Enfermedades Metabólicas and Research Direction of the Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, México; Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, México City, México
| | - Maria Teresa Tusié-Luna
- Unidad de Biología Molecular y Medicina Genómica, Instituto de Investigaciones Biomédicas UNAM Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, México
| | - Linda Liliana Muñoz-Hernandez
- Unidad de Investigación de Enfermedades Metabólicas del Instituto Nacional de Ciencias, Médicas, y Nutrición Salvador Zubirán, México City, México
| | - Miguel Herrera-Hernandez
- Surgery Direction of the Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, México
| | - Miryoung Lee
- Department of Epidemiology, The University of Texas Health Science Center at Houston School of Public Health, Brownsville Regional Campus, Brownsville, TX, USA
| | - Eric R Gamazon
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA; MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
| | - Nancy J Cox
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, USA; Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, USA; Institute for Precision Health at University of California, Los Angeles, Los Angeles, CA, USA
| | - Susan K Fried
- Obesity Research Center, Boston University School of Medicine, Boston, MA, USA; Diabetes Obesity and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Penny Gordon-Larsen
- Department of Nutrition, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ravi V Shah
- Vanderbilt Translational and Clinical Research Center, Cardiovascular Division, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Susan P Fisher-Hoch
- Department of Epidemiology, The University of Texas Health Science Center at Houston School of Public Health, Brownsville Regional Campus, Brownsville, TX, USA
| | - Joseph B McCormick
- Department of Epidemiology, The University of Texas Health Science Center at Houston School of Public Health, Brownsville Regional Campus, Brownsville, TX, USA
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Jennifer E Below
- Department of Medicine, Division of Genetic Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
11
|
Jiang W, Ding K, Yang M, Hu Z, Yue R. Exploring the Potential Effect of GLP1R Agonism on Common Aging-Related Diseases via Glucose Reduction: A Mendelian Randomization Study. J Gerontol A Biol Sci Med Sci 2025; 80:glaf007. [PMID: 39797952 DOI: 10.1093/gerona/glaf007] [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: 09/09/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND Glucagon-like peptide-1 receptor agonists (GLP1RAs) are widely used in managing type 2 diabetes mellitus and weight control. Their potential in treating aging-related diseases has been gaining attention in recent years. However, the long-term effects of GLP1RAs on these diseases have yet to be fully revealed. METHODS Using a genetic variant in the GLP1R gene to model the long-term effects of GLP1RAs, this Mendelian randomization (MR) study systematically explored potential causal associations between GLP1R agonism and 12 aging-related diseases and indicators. Genetic summary data sets used in this study were obtained from previous genome-wide association studies. RESULTS The primary MR analysis results suggested that GLP1R agonism was potentially positively causally associated with appendicular lean mass (Beta = 0.246, 95% confidence interval [CI] = 0.096-0.396), whole-body fat-free mass (Beta = 0.202, 95% CI = 0.048-0.355), and lung function (forced vital capacity [FVC]; Beta = 0.179, 95% CI = 0.152-0.205; p < .05). Additionally, a potential negative causal association was observed with myocardial infarction (odds ratio = 0.430, 95% CI = 0.249-0.745; p < .05). CONCLUSIONS The present MR study provides exploratory evidence suggesting potential causal associations between GLP1R agonism and appendicular lean mass, whole-body fat-free mass, lung function (FVC), and myocardial infarction. Given the exploratory nature of these findings and the limitations of the MR methodology, further research is needed to validate these results and investigate the underlying biological mechanisms.
Collapse
Affiliation(s)
- Wei Jiang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kaixi Ding
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Maoyi Yang
- Department of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhipeng Hu
- Department of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Rensong Yue
- Department of Clinical Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
12
|
Yang L, Sadler MC, Altman RB. Genetic association studies using disease liabilities from deep neural networks. Am J Hum Genet 2025; 112:675-692. [PMID: 39986278 PMCID: PMC11948217 DOI: 10.1016/j.ajhg.2025.01.019] [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: 09/05/2024] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/24/2025] Open
Abstract
The case-control study is a widely used method for investigating the genetic underpinnings of binary traits. However, long-term, prospective cohort studies often grapple with absent or evolving health-related outcomes. Here, we propose two methods, liability and meta, for conducting genome-wide association studies (GWASs) that leverage disease liabilities calculated from deep patient phenotyping. Analyzing 38 common traits in ∼300,000 UK Biobank participants, we identified an increased number of loci in comparison to the number identified by the conventional case-control approach, and there were high replication rates in larger external GWASs. Further analyses confirmed the disease specificity of the genetic architecture; the meta method demonstrated higher robustness when phenotypes were imputed with low accuracy. Additionally, polygenic risk scores based on disease liabilities more effectively predicted newly diagnosed cases in the 2022 dataset, which were controls in the earlier 2019 dataset. Our findings demonstrate that integrating high-dimensional phenotypic data into deep neural networks enhances genetic association studies while capturing disease-relevant genetic architecture.
Collapse
Affiliation(s)
- Lu Yang
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Computer Science, Stanford University, Stanford, CA 94305, USA.
| | - Marie C Sadler
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; University Center for Primary Care and Public Health, 1010 Lausanne, Switzerland; Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland
| | - Russ B Altman
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Genetics, Stanford University, Stanford, CA 94305, USA; Department of Medicine, Stanford University, Stanford, CA 94305, USA; Department of Computer Science, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
13
|
Li P, Ye H, Guo F, Zheng J, Shen W, Xie D, Shi S, Zhang Y, Fa Y, Zhao Z. Construction of cynomolgus monkey type 2 diabetes models by combining genetic prediction model with high-energy diet. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167616. [PMID: 39672349 DOI: 10.1016/j.bbadis.2024.167616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/01/2024] [Accepted: 12/02/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2D) is a significant health concern. Research using non-human primates, which develop T2D with similar symptoms and pancreatic changes as humans, is crucial but limited by long timelines and low success rates. RESULTS We targeted capture sequenced 61 normal and 81 T2D cynomolgus monkeys using a primer panel that captured 269 potential regulatory regions potentially associated with T2D in the cynomolgus monkey genome. 80 variants were identified to be associated with T2D and were used to construct a genetic prediction model. Among 8 machine learning algorithms tested, we found that the best prediction performance was achieve when the model using support vector machine with polynomial kernel as the machine learning algorithm (AUC = 0.933). Including age and sex in this model did not significantly improve the prediction performance. Using the genetic prediction model, we further screened 22 monkeys and found 13 were high risk while 9 were low risk. After feeding the 22 monkeys with high-energy food for 32 weeks, we found all the 9 low risk monkeys did not develop T2D while 4 out of 13 high risk monkeys (31 %) develop T2D. CONCLUSIONS This method greatly increased the success rate of establishing T2D monkey models while decreased the time needed compared to traditional methods. Therefore, we developed a new high-efficiency method to establish T2D monkey models by combining the genetic prediction model and high-energy diet, which will greatly contribute to the research on the clinical characteristics, pathogenesis, complications and potential new treatments.
Collapse
Affiliation(s)
- Ping Li
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Huahu Ye
- Academy of Military Medical Sciences, Beijing, China
| | - Feng Guo
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Jianhua Zheng
- Academy of Military Medical Sciences, Beijing, China
| | - Wenlong Shen
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Dejian Xie
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Shu Shi
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| | - Yan Zhang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China.
| | - Yunzhi Fa
- Academy of Military Medical Sciences, Beijing, China
| | - Zhihu Zhao
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, China
| |
Collapse
|
14
|
Martin SS, Aday AW, Allen NB, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Bansal N, Beaton AZ, Commodore-Mensah Y, Currie ME, Elkind MSV, Fan W, Generoso G, Gibbs BB, Heard DG, Hiremath S, Johansen MC, Kazi DS, Ko D, Leppert MH, Magnani JW, Michos ED, Mussolino ME, Parikh NI, Perman SM, Rezk-Hanna M, Roth GA, Shah NS, Springer MV, St-Onge MP, Thacker EL, Urbut SM, Van Spall HGC, Voeks JH, Whelton SP, Wong ND, Wong SS, Yaffe K, Palaniappan LP. 2025 Heart Disease and Stroke Statistics: A Report of US and Global Data From the American Heart Association. Circulation 2025; 151:e41-e660. [PMID: 39866113 DOI: 10.1161/cir.0000000000001303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
BACKGROUND The American Heart Association (AHA), in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, nutrition, sleep, and obesity) and health factors (cholesterol, blood pressure, glucose control, and metabolic syndrome) that contribute to cardiovascular health. The AHA Heart Disease and Stroke Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, brain health, complications of pregnancy, kidney disease, congenital heart disease, rhythm disorders, sudden cardiac arrest, subclinical atherosclerosis, coronary heart disease, cardiomyopathy, heart failure, valvular disease, venous thromboembolism, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The AHA, through its Epidemiology and Prevention Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States and globally to provide the most current information available in the annual Statistical Update with review of published literature through the year before writing. The 2025 AHA Statistical Update is the product of a full year's worth of effort in 2024 by dedicated volunteer clinicians and scientists, committed government professionals, and AHA staff members. This year's edition includes a continued focus on health equity across several key domains and enhanced global data that reflect improved methods and incorporation of ≈3000 new data sources since last year's Statistical Update. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
15
|
Liu J, Liu X, Rao R, Li W. TCF7L2 as a target of peripheral artery disease in patients with type 2 diabetes: A 2-sample Mendelian randomization and bioinformatics study. Medicine (Baltimore) 2025; 104:e41431. [PMID: 39960897 PMCID: PMC11835089 DOI: 10.1097/md.0000000000041431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 01/16/2025] [Indexed: 02/20/2025] Open
Abstract
This study examines the causal relationship between type 2 diabetes (T2D) and peripheral artery disease (PAD) and their potential mechanisms based on the analysis of the Gene Expression Omnibus database and 2-sample Mendelian randomization (MR). The first part involved a 2-sample MR study and a comprehensive meta-analysis. Differences in the results were assessed using inverse-variance weighting. Heterogeneity was examined using the Cochrane Q statistical test. The leave-one-out method was applied for sensitivity analysis. The potential horizontal pleiotropic effect was assessed using the MR-Egger intercept technique. The second part involved differential gene analysis and weighted gene coexpression network analysis. Subsequently, we overlapped and consolidated the results from the 2 parts to identify the key genes between them. MR analysis results suggested a statistically significant correlation between the incidence of PAD and T2D (odds ratio: 1.22, 95% confidence interval: 1.13-1.32, P = 3.74e-07). We anticipated a pivotal role for TCF7L2 in PAD and T2D. T2D was significantly associated with PAD risk. Simultaneously, the study deepened our understanding of the underlying mechanisms of both diseases, proposing TCF7L2 as a promising target.
Collapse
Affiliation(s)
- Jie Liu
- Department of Basic Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
- Department of Cardiology, Longli Hospital of Traditional Chinese Medicine, Qiannan, Guizhou, China
| | - XingDe Liu
- Department of Cardiology, Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Rui Rao
- Department of Endocrinology, Second Affiliated Hospital of Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| | - Wen Li
- Department of Basic Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China
| |
Collapse
|
16
|
Pan L, Liu Y, Huang C, Huang Y, Lin R, Wei K, Yao Y, Qin G, Yu Y. Association of accelerated phenotypic aging, genetic risk, and lifestyle with progression of type 2 diabetes: a prospective study using multi-state model. BMC Med 2025; 23:62. [PMID: 39901253 PMCID: PMC11792689 DOI: 10.1186/s12916-024-03832-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 12/18/2024] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Aging is a major risk factor for type 2 diabetes (T2D), but individuals of the same chronological age may vary in their biological aging rate. The associations of Phenotypic Age Acceleration (PhenoAgeAccel), a new accelerated biological aging indicator based on clinical chemistry biomarkers, with the risk of dynamic progression remain unclear. We aimed to assess these associations and examine whether these associations varied by genetic risk and lifestyle. METHODS We conducted a prospective cohort study that included 376,083 adults free of T2D and diabetes-related events at baseline in UK Biobank. PhenoAgeAccel > 0 and ≤ 0 were defined as biologically older and younger than chronological age. The outcomes of interest were incident T2D, diabetic complications, and mortality. Hazard ratios (HRs) with 95% confidence intervals (CIs) and population attributable fractions (PAFs) for these associations were calculated using multi-state model. RESULTS During a median follow-up of 13.7 years, 17,615 participants developed T2D, of whom, 4,524 subsequently developed complications, and 28,373 died. Being biologically older was associated with increased risks of transitions from baseline to T2D (HR 1.77, 95% CI 1.71-1.82; PAF 24.8 [95% CI 23.5-26.2]), from T2D to diabetic complications (1.10, 1.04-1.17; 4.4 [1.4-7.4]), from baseline to all-cause death (1.53, 1.49-1.57; 17.6 [16.6-18.6]), from T2D to all-cause death (1.14, 1.03-1.26; 7.4 [1.8-13.0]), and from diabetic complications to all-cause death (1.32, 1.15-1.51; 15.4 [7.5-23.2]) than being biologically younger. Additionally, participants with older biological age and high genetic risk had a higher risk of incident T2D (4.76,4.43-5.12;18.2 [17.5-19.0]) than those with younger biological age and low genetic risk. Compared with participants with younger biological age and healthy lifestyle, those with older biological age and unhealthy lifestyle had higher risks of transitions in the T2D trajectory, with HRs and PAFs ranging from 1.34 (1.16-1.55; 3.7 [1.8-5.6]) to 5.39 (5.01-5.79; 13.0 [12.4-13.6]). CONCLUSIONS PhenoAgeAccel was consistently associated with an increased risk of all transitions in T2D progression. It has the potential to be combined with genetic risk to identify early T2D incidence risk and may guide interventions throughout T2D progression while tracking their effectiveness.
Collapse
Affiliation(s)
- Lulu Pan
- Department of Biostatistics, Key Laboratory of Public Health Safety of Ministry of Education, NHC Key Laboratory for Health Technology Assessment, School of Public Health, Fudan University, 138 Yi Xue Yuan Road, Shanghai, 200032, China
| | - Yahang Liu
- Department of Biostatistics, Key Laboratory of Public Health Safety of Ministry of Education, NHC Key Laboratory for Health Technology Assessment, School of Public Health, Fudan University, 138 Yi Xue Yuan Road, Shanghai, 200032, China
| | - Chen Huang
- Department of Biostatistics, Key Laboratory of Public Health Safety of Ministry of Education, NHC Key Laboratory for Health Technology Assessment, School of Public Health, Fudan University, 138 Yi Xue Yuan Road, Shanghai, 200032, China
| | - Yifang Huang
- Department of Biostatistics, Key Laboratory of Public Health Safety of Ministry of Education, NHC Key Laboratory for Health Technology Assessment, School of Public Health, Fudan University, 138 Yi Xue Yuan Road, Shanghai, 200032, China
| | - Ruilang Lin
- Department of Biostatistics, Key Laboratory of Public Health Safety of Ministry of Education, NHC Key Laboratory for Health Technology Assessment, School of Public Health, Fudan University, 138 Yi Xue Yuan Road, Shanghai, 200032, China
| | - Kecheng Wei
- Department of Biostatistics, Key Laboratory of Public Health Safety of Ministry of Education, NHC Key Laboratory for Health Technology Assessment, School of Public Health, Fudan University, 138 Yi Xue Yuan Road, Shanghai, 200032, China
| | - Ye Yao
- Department of Biostatistics, Key Laboratory of Public Health Safety of Ministry of Education, NHC Key Laboratory for Health Technology Assessment, School of Public Health, Fudan University, 138 Yi Xue Yuan Road, Shanghai, 200032, China
| | - Guoyou Qin
- Department of Biostatistics, Key Laboratory of Public Health Safety of Ministry of Education, NHC Key Laboratory for Health Technology Assessment, School of Public Health, Fudan University, 138 Yi Xue Yuan Road, Shanghai, 200032, China.
- Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China.
| | - Yongfu Yu
- Department of Biostatistics, Key Laboratory of Public Health Safety of Ministry of Education, NHC Key Laboratory for Health Technology Assessment, School of Public Health, Fudan University, 138 Yi Xue Yuan Road, Shanghai, 200032, China.
| |
Collapse
|
17
|
Korvyakova Y, Azarova I, Klyosova E, Postnikova M, Makarenko V, Bushueva O, Solodilova M, Polonikov A. The link between the ANPEP gene and type 2 diabetes mellitus may be mediated by the disruption of glutathione metabolism and redox homeostasis. Gene 2025; 935:149050. [PMID: 39489227 DOI: 10.1016/j.gene.2024.149050] [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: 05/15/2024] [Revised: 10/02/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024]
Abstract
Aminopeptidase N (ANPEP), a membrane-associated ectoenzyme, has been identified as a susceptibility gene for type 2 diabetes (T2D) by genome-wide association and transcriptome studies; however, the mechanisms by which this gene contributes to disease pathogenesis remain unclear. The aim of this study was to determine the comprehensive contribution of ANPEP polymorphisms to T2D risk and annotate the underlying mechanisms. A total of 3206 unrelated individuals including 1579 T2D patients and 1627 controls were recruited for the study. Twenty-three common functional single nucleotide polymorphisms (SNP) of ANPEP were genotyped by the MassArray-4 system. Six polymorphisms, rs11073891, rs12898828, rs12148357, rs9920421, rs7111, and rs25653, were found to be associated with type 2 diabetes (Pperm ≤ 0.05). Common haplotype rs9920421G-rs4932143G-rs7111T was strongly associated with increased risk of T2D (Pperm = 5.9 × 10-12), whereas two rare haplotypes such as rs9920421G-rs4932143C-rs7111T (Pperm = 6.5 × 10-40) and rs12442778A-rs12898828A-rs6496608T-rs11073891C (Pperm = 1.0 × 10-7) possessed strong protection against disease. We identified 38 and 109 diplotypes associated with T2D risk in males and females, respectively (FDR ≤ 0.05). ANPEP polymorphisms showed associations with plasma levels of fasting blood glucose, aspartate aminotransferase, total protein and glutathione (P < 0.05), and several haplotypes were strongly associated with the levels of reactive oxygen species and uric acid (P < 0.0001). A deep literature analysis has facilitated the formulation of a hypothesis proposing that increased plasma levels of ANPEP as well as liver enzymes such as aspartate aminotransferase, alanine aminotransferase and gammaglutamyltransferase serve as an adaptive response directed towards the restoration of glutathione deficiency in diabetics by stimulating the production of amino acid precursors for glutathione biosynthesis.
Collapse
Affiliation(s)
- Yaroslava Korvyakova
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation; Research Centre for Medical Genetics, 1 Moskvorechie St., Moscow 115522, Russian Federation.
| | - Iuliia Azarova
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation; Department of Biological Chemistry, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russian Federation.
| | - Elena Klyosova
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation; Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russian Federation.
| | - Maria Postnikova
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation.
| | - Victor Makarenko
- Laboratory of Biochemical Genetics and Metabolomics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation
| | - Olga Bushueva
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russian Federation; Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation.
| | - Maria Solodilova
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russian Federation.
| | - Alexey Polonikov
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, 3 Karl Marx Street, Kursk 305041, Russian Federation; Laboratory of Statistical Genetics and Bioinformatics, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, 18 Yamskaya St., Kursk 305041, Russian Federation.
| |
Collapse
|
18
|
Abad-González ÁL, Veses S, Argente Pla M, Civera M, García-Malpartida K, Sánchez C, Artero A, Palmas F, Perelló E, Salom C, Yun Wu Xiong N, Joaquim C. Medical Nutrition Therapy and Physical Exercise for Acute and Chronic Hyperglycemic Patients with Sarcopenia. Nutrients 2025; 17:499. [PMID: 39940355 PMCID: PMC11820730 DOI: 10.3390/nu17030499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/21/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
A wide range of factors contribute to the overlap of hyperglycemia-acute or chronic-and sarcopenia, as well as their associated adverse consequences, which can lead to impaired physical function, reduced quality of life, and increased mortality risk. These factors include malnutrition (both overnutrition and undernutrition) and low levels of physical activity. Hyperglycemia and sarcopenia are interconnected through a vicious cycle of events that mutually reinforce and worsen each other. To explore this association, our review compiles evidence on: (i) the impact of hyperglycemia on motor and muscle function, with a focus on the mechanisms underlying biochemical changes in the muscles of individuals with or at risk of diabetes and sarcopenia; (ii) the importance of the clinical assessment and control of sarcopenia under hyperglycemic conditions; and (iii) the potential benefits of medical nutrition therapy and increased physical activity as muscle-targeted treatments for this population. Based on the reviewed evidence, we conclude that a regular intake of key functional nutrients, together with structured and supervised resistance and/or aerobic physical activity, can help maintain euglycemia and improve muscle status in all patients with hyperglycemia and sarcopenia.
Collapse
Affiliation(s)
- Ángel Luis Abad-González
- Endocrinology and Nutrition Department, Hospital General Universitario Dr. Balmis, 03010 Alicante, Spain;
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), 03010 Alicante, Spain
| | - Silvia Veses
- Endocrinology and Nutrition Department, Hospital Universitario Doctor Peset, 46017 Valencia, Spain; (S.V.); (K.G.-M.); (C.S.)
| | - María Argente Pla
- Endocrinology Department, Hospital Universitari i Politècnic La Fe, 46026 Valencia, Spain;
| | - Miguel Civera
- Endocrinology and Nutrition Department, University Clinical Hospital, Valencia, INCLIVA Biomedical Research Institute, 46010 Valencia, Spain;
| | - Katherine García-Malpartida
- Endocrinology and Nutrition Department, Hospital Universitario Doctor Peset, 46017 Valencia, Spain; (S.V.); (K.G.-M.); (C.S.)
- School of Health Sciences, Universidad Cardenal Herrera-CEU, CEU Universities, Calle Grecia 31, 12006 Castellón, Spain
| | - Carlos Sánchez
- Endocrinology and Nutrition Department, Consorcio Hospital General Universitario de Valencia, Departamento de Medicina, University of Valencia, 46016 Valencia, Spain; (C.S.); (A.A.)
| | - Ana Artero
- Endocrinology and Nutrition Department, Consorcio Hospital General Universitario de Valencia, Departamento de Medicina, University of Valencia, 46016 Valencia, Spain; (C.S.); (A.A.)
| | - Fiorella Palmas
- Endocrinology Department, Hospital Universitari Vall d’Hebron, 08035 Barcelona, Spain;
| | - Eva Perelló
- Endocrinology Department, Hospital Universitario San Juan de Alicante, 03550 Alicante, Spain;
| | - Christian Salom
- Endocrinology and Nutrition Department, Hospital Universitario Doctor Peset, 46017 Valencia, Spain; (S.V.); (K.G.-M.); (C.S.)
| | - Ning Yun Wu Xiong
- Endocrinology Department, Hospital Clínico Universitario de Valencia, 46010 Valencia, Spain;
| | - Clara Joaquim
- Endocrinology Department, Hospital Universitari Germans Trias i Pujol, 08916 Badalona, Spain
| |
Collapse
|
19
|
Yang C, Ji L, Han X. Low C-Reactive Protein Alleles in Hepatocyte Nuclear Factor 1A Are Associated With an Increased Risk of Cardiovascular Disease. J Clin Endocrinol Metab 2025; 110:592-600. [PMID: 39210612 DOI: 10.1210/clinem/dgae602] [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: 05/04/2024] [Revised: 07/10/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
CONTEXT Rare variants in HNF1A cause both maturity onset diabetes of the young 3 (HNF1A-MODY) and reduced serum C-reactive protein (CRP) levels. Common variants of HNF1A are associated with serum CRP and type 2 diabetes mellitus (T2DM), but inconsistently with cardiovascular disease (CVD). OBJECTIVE Our study aimed to investigate the association of low CRP alleles in HNF1A with CVD and indirectly evaluate the CVD risk of HNF1A-MODY patients because of unavailability of enough cases to study their clinical outcomes. METHODS A literature search was performed using PubMed, Embase, and Cochrane Library databases from inception to December 2023. All relevant studies concerning the association of HNF1A with CRP, CVD, lipids, and T2DM were included. Odds ratios (ORs), 95% CIs, and study characteristics were extracted. RESULTS Three common coding variants of HNF1A (rs1169288, rs2464196, and rs1169289) were examined. The minor alleles of these variants correlated with low CRP levels (OR 0.89; 95% CI, 0.86-0.91; OR 0.89; 95% CI, 0.88-0.91; OR 0.89; 95% CI, 0.88-0.91, respectively). Their low CRP alleles were associated with increased risk of CVD (OR 1.03; 95% CI, 1.03-1.04), higher low-density lipoprotein cholesterol levels (OR 1.07; 95% CI, 1.04-1.10), and elevated risk of T2DM (OR 1.04; 95%, CI 1.01-1.08). CONCLUSION Our study revealed an association between low CRP alleles in HNF1A and a high CVD risk, which indicated that antidiabetic drugs with CV benefits such as glucagon-like peptide-1 receptor agonists should be recommended as a first-line choice for HNF1A-MODY.
Collapse
Affiliation(s)
- Chaochao Yang
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing 100044, China
| | - Linong Ji
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing 100044, China
| | - Xueyao Han
- Department of Endocrinology and Metabolism, Peking University People's Hospital, Peking University Diabetes Center, Beijing 100044, China
| |
Collapse
|
20
|
Murtaza G, Riaz S, Zafar M, Ahsan Raza M, Kaleem I, Imran H, Al-Harbi AT, Sabouri A, Asim Niaz T, Bashir S. Examining the growing challenge: Prevalence of diabetes in young adults (Review). MEDICINE INTERNATIONAL 2025; 5:2. [PMID: 39563945 PMCID: PMC11571047 DOI: 10.3892/mi.2024.201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 10/14/2024] [Indexed: 11/21/2024]
Abstract
Diabetes is rapidly spreading worldwide, affecting millions of individuals. Therefore, it is crucial to have a comprehensive understanding of its complications. The present review discusses the complex subject of diabetes, including the type 1 and type 2 variants. Geographical and population differences highlight the importance of targeted therapies and personalized management strategies. Ongoing research aims to identify the causes and treatment strategies for this disease. Preventive interventions, lifestyle changes and public awareness campaigns are all vital components of diabetes management. Collaboration between the general public and health departments is essential for effective prevention. Early intervention and global management strategies are necessary to reduce the significant impact on healthcare systems. A comprehensive plan from health care departments is required to address the issues caused by diabetes and minimize its effects on individuals and communities worldwide. The present review outlines specific measures which can be used to combat the spread of diabetes for a healthier future world.
Collapse
Affiliation(s)
- Ghulam Murtaza
- Department of Zoology, University of Gujrat, Gujrat, Punjab 50700, Pakistan
| | - Samavia Riaz
- Department of Zoology, University of Gujrat, Gujrat, Punjab 50700, Pakistan
| | - Maria Zafar
- Department of Zoology, University of Gujrat, Gujrat, Punjab 50700, Pakistan
| | | | - Imdad Kaleem
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad 45550, Pakistan
| | - Hadia Imran
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad 45550, Pakistan
| | - Aryam T Al-Harbi
- Department of Medicine, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 34212, Kingdom of Saudi Arabia
| | - Ali Sabouri
- Department of Oncology, Mount Vernon Cancer Centre, East and North Hertfordshire NHS Trust, Northwood HA6 2RN, UK
| | - Talha Asim Niaz
- Faculty of Medicine, University of Southampton, Southampton, SO17 1BJ, UK
| | - Shahid Bashir
- Department of Neuroscience, Neuroscience Center, King Fahad Specialist Hospital Dammam, Dammam 31444, Kingdom of Saudi Arabia
| |
Collapse
|
21
|
Chiang CW, Chou YH, Huang CN, Lu WY, Liaw YP. Gender-specific genetic influence of rs1111875 on diabetes risk: Insights from the Taiwan biobank study. J Diabetes Investig 2025; 16:36-42. [PMID: 39555857 DOI: 10.1111/jdi.14359] [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: 07/14/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/19/2024] Open
Abstract
BACKGROUND This study investigates the gender-specific genetic influence of the single nucleotide polymorphism (SNP) rs1111875 on diabetes risk within the Taiwanese population using data from the Taiwan Biobank. Diabetes mellitus, particularly type 2 diabetes (T2D), is influenced by genetic factors, and the rs1111875 SNP near the hematopoietically expressed homeobox (HHEX) gene has been linked to T2D susceptibility. METHODS The study included 69,272 participants after excluding those from arsenic-polluted areas and those with incomplete data. Logistic regression models were used for analyses. RESULTS The analyses revealed that the CT genotype of rs1111875 was associated with an increased risk of diabetes (OR = 1.092, 95% CI = 1.030-1.157, P = 0.003), as was the TT genotype (OR = 1.280, 95% CI = 1.165-1.407, P < 0.001). The effect was more pronounced in women (CT: OR = 1.118, 95% CI = 1.036-1.207, P = 0.004; TT: OR = 1.404, 95% CI = 1.243-1.585, P < 0.001). Men exhibited a higher overall risk of diabetes (OR = 1.565, 95% CI = 1.445-1.694, P < 0.001) and had a higher prevalence (12.71% vs 7.80%, P < 0.001) compared to women. CONCLUSIONS The findings underscore the importance of considering gender differences in genetic studies of diabetes and suggest that personalized diabetes management strategies should account for both genetic and gender-specific risk factors. This research contributes to the broader understanding of genetic determinants of diabetes and their interaction with gender, aiming to enhance personalized healthcare strategies for diabetes prevention and treatment.
Collapse
Affiliation(s)
- Chih-Wei Chiang
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung City, Taiwan
- College of Health Care and Management, Chung Shan Medical University, Taichung City, Taiwan
- Department of Information Communications, Chinese Culture University, Taipei City, Taiwan
| | - Ying-Hsiang Chou
- Department of Radiation Oncology, Chung Shan Medical University Hospital, Taichung, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- School of Medical Imaging and Radiological Sciences, Chung Shan Medical University, Taichung, Taiwan
| | - Chien-Ning Huang
- Department of Internal Medicine, Chung Shan Medical University Hospital, Taichung City, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung City, Taiwan
| | - Wen-Yu Lu
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung City, Taiwan
| | - Yung-Po Liaw
- Department of Public Health and Institute of Public Health, Chung Shan Medical University, Taichung City, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung City, Taiwan
- Department of Medical Imaging, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
22
|
Benner C, Mahajan A, Pirinen M. Refining fine-mapping: Effect sizes and regional heritability. PLoS Genet 2025; 21:e1011480. [PMID: 39787248 PMCID: PMC11753704 DOI: 10.1371/journal.pgen.1011480] [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: 04/22/2024] [Revised: 01/22/2025] [Accepted: 11/01/2024] [Indexed: 01/12/2025] Open
Abstract
Recent statistical approaches have shown that the set of all available genetic variants explains considerably more phenotypic variance of complex traits and diseases than the individual variants that are robustly associated with these phenotypes. However, rapidly increasing sample sizes constantly improve detection and prioritization of individual variants driving the associations between genomic regions and phenotypes. Therefore, it is useful to routinely estimate how much phenotypic variance the detected variants explain for each region by taking into account the correlation structure of variants and the uncertainty in their causal status. Here we extend the FINEMAP software to estimate the effect sizes and regional heritability under the probabilistic model that assumes a handful of causal variants per region. Using the UK Biobank (UKB) data to simulate genomic regions, we demonstrate that FINEMAP provides higher precision and enables more detailed decomposition of regional heritability into individual variants than the variance component model implemented in BOLT or the fixed-effect model implemented in HESS, particularly when there are only a few causal variants in the fine-mapped region. Using data from 2,940 plasma proteins from the UKB study, we observed that on average FINEMAP identified 2.5 causal variants at an association signal and captured 36% more regional heritability than the variant with the lowest P-value. We estimate that in genomic regions with notable contribution to the total heritability, FINEMAP captures on average 13% and 40% more heritability than BOLT and HESS respectively. Our analysis shows how FINEMAP, BOLT and HESS relate to each other in cases where inference of a variant-level picture of the regional genetic architecture is possible.
Collapse
Affiliation(s)
- Christian Benner
- Genentech, South San Francisco, California, United States of America
| | - Anubha Mahajan
- Genentech, South San Francisco, California, United States of America
| | - Matti Pirinen
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Department of Mathematics and Statistics, University of Helsinki, Helsinki, Finland
| |
Collapse
|
23
|
Ding K, Qin X, Wang H, Wang K, Kang X, Yu Y, Liu Y, Gong H, Wu T, Chen D, Hu Y, Wang T, Wu Y. Identification of shared genetic etiology of cardiovascular and cerebrovascular diseases through common cardiometabolic risk factors. Commun Biol 2024; 7:1703. [PMID: 39730871 DOI: 10.1038/s42003-024-07417-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 12/18/2024] [Indexed: 12/29/2024] Open
Abstract
Cardiovascular diseases (CVDs) and cerebrovascular diseases (CeVDs) are closely related vascular diseases, sharing common cardiometabolic risk factors (RFs). Although pleiotropic genetic variants of these two diseases have been reported, their underlying pathological mechanisms are still unclear. Leveraging GWAS summary data and using genetic correlation, pleiotropic variants identification, and colocalization analyses, we identified 11 colocalized loci for CVDs-CeVDs-BP (blood pressure), CVDs-CeVDs-LIP (lipid traits), and CVDs-CeVDs-cIMT (carotid intima-media thickness) triplets. No shared causal loci were found for CVDs-CeVDs-T2D (type 2 diabetes) or CVDs-CeVDs-BMI (body mass index) triplets. The 11 loci were mapped to 12 genes, namely CASZ1, CDKN1A, TWIST1, CDKN2B, ABO, SWAP70, SH2B3, LRCH1, FES, GOSR2, RPRML, and LDLR, where both GOSR2 and RPRML were mapped to one locus. They were enriched in pathways related to cellular response to external stimulus and regulation of the phosphate metabolic process and were highly expressed in endothelial cells, epithelial cells, and smooth muscle cells. Multi-omics analysis revealed methylation of two genes (CASZ1 and LRCH1) may play a causal role in the genetic pleiotropy. Notably, these pleiotropic loci are highly enriched in the targets of antihypertensive drugs, which further emphasizes the role of the blood pressure regulation pathway in the shared etiology of CVDs and CeVDs.
Collapse
Affiliation(s)
- Kexin Ding
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Xueying Qin
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China.
| | - Huairong Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Kun Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Xiaoying Kang
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Neurology, Brigham and Women's Hospital & Harvard Medical School, Boston, MA, USA
| | - Yao Yu
- Department of Neurology, Peking University People's Hospital, Beijing, China
| | - Yang Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Haiying Gong
- Fangshan District Center for Disease Control and Prevention, Beijing, China
| | - Tao Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Dafang Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Yonghua Hu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China
| | - Tao Wang
- Department of Epidemiology and Population Health, Yeshiva University Albert Einstein College of Medicine, Bronx, NY, USA
| | - Yiqun Wu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University; Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of Education, Beijing, China.
| |
Collapse
|
24
|
Thompson JR, Nelson ED, Tippani M, Ramnauth AD, Divecha HR, Miller RA, Eagles NJ, Pattie EA, Kwon SH, Bach SV, Kaipa UM, Yao J, Hou C, Kleinman JE, Collado-Torres L, Han S, Maynard KR, Hyde TM, Martinowich K, Page SC, Hicks SC. An integrated single-nucleus and spatial transcriptomics atlas reveals the molecular landscape of the human hippocampus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.26.590643. [PMID: 38712198 PMCID: PMC11071618 DOI: 10.1101/2024.04.26.590643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
The hippocampus contains many unique cell types, which serve the structure's specialized functions, including learning, memory and cognition. These cells have distinct spatial organization, morphology, physiology, and connectivity, highlighting the importance of transcriptome-wide profiling strategies that retain cytoarchitectural organization. Here, we generated spatially-resolved transcriptomics (SRT) and single-nucleus RNA-sequencing (snRNA-seq) data from adjacent tissue sections of the anterior human hippocampus in ten adult neurotypical donors to define molecular profiles for hippocampal cell types and spatial domains. Using non-negative matrix factorization (NMF) and label transfer, we integrated these data by defining gene expression patterns within the snRNA-seq data and inferring their expression in the SRT data. We identified NMF patterns that captured transcriptional variation across neuronal cell types and indicated that the response of excitatory and inhibitory postsynaptic specializations were prioritized in different SRT spatial domains. We used the NMF and label transfer approach to leverage existing rodent datasets, identifying patterns of activity-dependent transcription and subpopulations of dentate gyrus granule cells in our SRT dataset that may be predisposed to participate in learning and memory ensembles. Finally, we characterized the spatial organization of NMF patterns corresponding to non-cornu ammonis pyramidal neurons and identified snRNA-seq clusters mapping to distinct regions of the retrohippocampus, to three subiculum layers, and to a population of presubiculum neurons. To make this comprehensive molecular atlas accessible to the scientific community, both raw and processed data are freely available, including through interactive web applications.
Collapse
Affiliation(s)
- Jacqueline R. Thompson
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Erik D. Nelson
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Cellular and Molecular Medicine Graduate Program, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Madhavi Tippani
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Anthony D. Ramnauth
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Heena R. Divecha
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Ryan A. Miller
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Nicholas J. Eagles
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Elizabeth A. Pattie
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Sang Ho Kwon
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Biochemistry, Cellular, and Molecular Biology Graduate Program, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Svitlana V. Bach
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Uma M. Kaipa
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Jianing Yao
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Christine Hou
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Joel E. Kleinman
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Leonardo Collado-Torres
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Shizhong Han
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Genetic Medicine, Johns Hopkins School of Medicine, MD, USA
| | - Kristen R. Maynard
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Thomas M. Hyde
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Genetic Medicine, Johns Hopkins School of Medicine, MD, USA
- Johns Hopkins Kavli Neuroscience Discovery Institute, Baltimore, MD, USA
| | - Stephanie C. Page
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Stephanie C. Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
25
|
Zhao Y, Zhou R, Mu Z, Carbonetto P, Zhong X, Xie B, Luo K, Cham CM, Koval J, He X, Dahl AW, Liu X, Chang EB, Basu A, Pott S. Cell-type-resolved chromatin accessibility in the human intestine identifies complex regulatory programs and clarifies genetic associations in Crohn's disease. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.10.24318718. [PMID: 39711713 PMCID: PMC11661348 DOI: 10.1101/2024.12.10.24318718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Crohn's disease (CD) is a complex inflammatory bowel disease resulting from an interplay of genetic, microbial, and environmental factors. Cell-type-specific contributions to CD etiology and genetic risk are incompletely understood. Here we built a comprehensive atlas of cell-type- resolved chromatin accessibility comprising 557,310 candidate cis-regulatory elements (cCREs) in terminal ileum and ascending colon from patients with active and inactive CD and healthy controls. Using this atlas, we identified cell-type-, anatomic location-, and context-specific cCREs and characterized the regulatory programs underlying inflammatory responses in the intestinal mucosa of CD patients. Genetic variants that disrupt binding motifs of cell-type-specific transcription factors significantly affected chromatin accessibility in specific mucosal cell types. We found that CD heritability is primarily enriched in immune cell types. However, using fine- mapped non-coding CD variants we identified 29 variants located within cCREs several of which were accessible in epithelial and stromal cells implicating cell types from additional lineages in mediating CD risk in some loci. Our atlas provides a comprehensive resource to study gene regulatory effects in CD and health, and highlights the cellular complexity underlying CD risk.
Collapse
|
26
|
Liu S, Zhu J, Zhong H, Wu C, Xue H, Darst BF, Guo X, Durda P, Tracy RP, Liu Y, Johnson WC, Taylor KD, Manichaikul AW, Goodarzi MO, Gerszten RE, Clish CB, Chen YDI, Highland H, Haiman CA, Gignoux CR, Lange L, Conti DV, Raffield LM, Wilkens L, Marchand LL, North KE, Young KL, Loos RJ, Buyske S, Matise T, Peters U, Kooperberg C, Reiner AP, Yu B, Boerwinkle E, Sun Q, Rooney MR, Echouffo-Tcheugui JB, Daviglus ML, Qi Q, Mancuso N, Li C, Deng Y, Manning A, Meigs JB, Rich SS, Rotter JI, Wu L. Identification of proteins associated with type 2 diabetes risk in diverse racial and ethnic populations. Diabetologia 2024; 67:2754-2770. [PMID: 39349773 PMCID: PMC11963907 DOI: 10.1007/s00125-024-06277-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 07/16/2024] [Indexed: 11/29/2024]
Abstract
AIMS/HYPOTHESIS Several studies have reported associations between specific proteins and type 2 diabetes risk in European populations. To better understand the role played by proteins in type 2 diabetes aetiology across diverse populations, we conducted a large proteome-wide association study using genetic instruments across four racial and ethnic groups: African; Asian; Hispanic/Latino; and European. METHODS Genome and plasma proteome data from the Multi-Ethnic Study of Atherosclerosis (MESA) study involving 182 African, 69 Asian, 284 Hispanic/Latino and 409 European individuals residing in the USA were used to establish protein prediction models by using potentially associated cis- and trans-SNPs. The models were applied to genome-wide association study summary statistics of 250,127 type 2 diabetes cases and 1,222,941 controls from different racial and ethnic populations. RESULTS We identified three, 44 and one protein associated with type 2 diabetes risk in Asian, European and Hispanic/Latino populations, respectively. Meta-analysis identified 40 proteins associated with type 2 diabetes risk across the populations, including well-established as well as novel proteins not yet implicated in type 2 diabetes development. CONCLUSIONS/INTERPRETATION Our study improves our understanding of the aetiology of type 2 diabetes in diverse populations. DATA AVAILABILITY The summary statistics of multi-ethnic type 2 diabetes GWAS of MVP, DIAMANTE, Biobank Japan and other studies are available from The database of Genotypes and Phenotypes (dbGaP) under accession number phs001672.v3.p1. MESA genetic, proteome and covariate data can be accessed through dbGaP under phs000209.v13.p3. All code is available on GitHub ( https://github.com/Arthur1021/MESA-1K-PWAS ).
Collapse
Affiliation(s)
- Shuai Liu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawai'i Cancer Center, University of Hawai'i at Mānoa, Honolulu, HI, USA
| | - Jingjing Zhu
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawai'i at Mānoa, Honolulu, HI, USA
| | - Hua Zhong
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawai'i Cancer Center, University of Hawai'i at Mānoa, Honolulu, HI, USA
| | - Chong Wu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Haoran Xue
- Division of Biostatistics, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Burcu F Darst
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Peter Durda
- Laboratory for Clinical Biochemistry Research, University of Vermont, Burlington, VT, USA
| | - Russell P Tracy
- Laboratory for Clinical Biochemistry Research, University of Vermont, Burlington, VT, USA
| | - Yongmei Liu
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - W Craig Johnson
- Collaborative Health Studies Coordinating Center, University of Washington, Seattle, WA, USA
| | - Kent D Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Ani W Manichaikul
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, USA
| | - Mark O Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Robert E Gerszten
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Clary B Clish
- Metabolomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Heather Highland
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Christopher A Haiman
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Christopher R Gignoux
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Leslie Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - David V Conti
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Laura M Raffield
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Lynne Wilkens
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawai'i Cancer Center, University of Hawai'i at Mānoa, Honolulu, HI, USA
| | - Loïc Le Marchand
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawai'i Cancer Center, University of Hawai'i at Mānoa, Honolulu, HI, USA
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kristin L Young
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ruth J Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Steve Buyske
- Department of Statistics, Rutgers University, Piscataway, NJ, USA
| | - Tara Matise
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | | | - Bing Yu
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Quan Sun
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mary R Rooney
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Justin B Echouffo-Tcheugui
- Division of Endocrinology, Diabetes and Metabolism, Johns Hopkins Bayview Medical Center, Baltimore, MD, USA
| | - Martha L Daviglus
- Institute for Minority Health Research, University of Illinois at Chicago, Chicago, IL, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Nicholas Mancuso
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Changwei Li
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Youping Deng
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawai'i at Mānoa, Honolulu, HI, USA
| | - Alisa Manning
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, MA, USA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - James B Meigs
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Stephen S Rich
- Department of Genome Sciences, University of Virginia, Charlottesville, VA, USA
| | - Jerome I Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Lang Wu
- Cancer Epidemiology Division, Population Sciences in the Pacific Program, University of Hawai'i Cancer Center, University of Hawai'i at Mānoa, Honolulu, HI, USA.
| |
Collapse
|
27
|
Fu L, Cheng H, Xiong J, Xiao P, Shan X, Li Y, Li Y, Zhao X, Mi J. Mediating role of inflammatory biomarkers in the causal effect of body composition on glycaemic traits and type 2 diabetes. Diabetes Obes Metab 2024; 26:5444-5454. [PMID: 39228266 DOI: 10.1111/dom.15923] [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: 07/16/2024] [Revised: 08/20/2024] [Accepted: 08/20/2024] [Indexed: 09/05/2024]
Abstract
OBJECTIVE The aim was to investigate the mediating role of inflammatory biomarkers in the causal effect of body composition on glycaemic traits and type 2 diabetes. METHODS A retrospective observational study and a Mendelian randomization (MR) study were used. Observational analyses were performed using data from 4717 Chinese children and adolescents aged 6-18 years who underwent dual-energy X-ray absorptiometry for body composition. MR analyses were based on summary statistics from UK Biobank, deCODE2021, Meta-Analysis of Glucose and Insulin-Related Traits Consortium (MAGIC) and other large consortiums. Inflammatory biomarkers included leptin, adiponectin, osteocalcin, fibroblast growth factor 23 (FGF23) and parathyroid hormone (PTH). RESULTS In a retrospective observational study, increased fat mass had a positive effect on homeostasis model assessment of insulin resistance (HOMA-IR) and homeostasis model assessment of pancreatic beta cell function (HOMA-β) through FGF23, whereas fat-free mass produced the opposite effects. PTH and osteocalcin played significant roles in the association of fat mass and fat-free mass with fasting glucose, fasting insulin and HOMA-IR (all p < 0.05). Mediation MR results indicated that childhood body mass index affected glycaemic traits through leptin and adiponectin. There existed a causal effect of fat-free mass on type 2 diabetes via FGF23 (indirect effect: OR [odds ratio]: 1.14 [95% CI, confidence interval: 1.01-1.28]) and adiponectin (OR: 0.85 [95% CI: 0.77-0.93]). Leptin mediated the causal association of fat mass (indirect effect: β: -0.05 [95% CI: -0.07, -0.02]) and fat-free mass (β: 0.03 [95% CI: 0.01, 0.04]) with fasting glucose. CONCLUSIONS Our findings suggest that different body compositions have differential influences on glycaemic traits and type 2 diabetes through distinct inflammatory biomarkers. The findings may be helpful in tailoring management of body composition based on inflammatory biomarkers with different glycaemic statuses.
Collapse
Affiliation(s)
- Liwan Fu
- Center for Non-Communicable Disease Management, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Hong Cheng
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| | - Jingfan Xiong
- Child and Adolescent Chronic Disease Prevention and Control Department, Shenzhen Center for Chronic Disease Control, Shenzhen, China
| | - Pei Xiao
- Center for Non-Communicable Disease Management, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xinying Shan
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| | - Yanyan Li
- Child and Adolescent Chronic Disease Prevention and Control Department, Shenzhen Center for Chronic Disease Control, Shenzhen, China
| | - Yan Li
- Child and Adolescent Chronic Disease Prevention and Control Department, Shenzhen Center for Chronic Disease Control, Shenzhen, China
| | - Xiaoyuan Zhao
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| | - Jie Mi
- Center for Non-Communicable Disease Management, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
- Key Laboratory of Major Diseases in Children, Ministry of Education, Beijing, China
| |
Collapse
|
28
|
Sullivan KA, Lane M, Cashman M, Miller JI, Pavicic M, Walker AM, Cliff A, Romero J, Qin X, Mullins N, Docherty A, Coon H, Ruderfer DM, Garvin MR, Pestian JP, Ashley-Koch AE, Beckham JC, McMahon B, Oslin DW, Kimbrel NA, Jacobson DA, Kainer D. Analyses of GWAS signal using GRIN identify additional genes contributing to suicidal behavior. Commun Biol 2024; 7:1360. [PMID: 39433874 PMCID: PMC11494055 DOI: 10.1038/s42003-024-06943-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 09/23/2024] [Indexed: 10/23/2024] Open
Abstract
Genome-wide association studies (GWAS) identify genetic variants underlying complex traits but are limited by stringent genome-wide significance thresholds. We present GRIN (Gene set Refinement through Interacting Networks), which increases confidence in the expanded gene set by retaining genes strongly connected by biological networks when GWAS thresholds are relaxed. GRIN was validated on both simulated interrelated gene sets as well as multiple GWAS traits. From multiple GWAS summary statistics of suicide attempt, a complex phenotype, GRIN identified additional genes that replicated across independent cohorts and retained biologically interrelated genes despite a relaxed significance threshold. We present a conceptual model of how these retained genes interact through neurobiological pathways that may influence suicidal behavior, and identify existing drugs associated with these pathways that would not have been identified under traditional GWAS thresholds. We demonstrate GRIN's utility in boosting GWAS results by increasing the number of true positive genes identified from GWAS results.
Collapse
Affiliation(s)
- Kyle A Sullivan
- Computational and Predictive Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Matthew Lane
- The Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee Knoxville, Knoxville, TN, USA
| | - Mikaela Cashman
- Computational and Predictive Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory Berkeley, California, CA, USA
| | - J Izaak Miller
- Computational and Predictive Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Mirko Pavicic
- Computational and Predictive Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - Angelica M Walker
- The Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee Knoxville, Knoxville, TN, USA
| | - Ashley Cliff
- The Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee Knoxville, Knoxville, TN, USA
| | - Jonathon Romero
- The Bredesen Center for Interdisciplinary Research and Graduate Education, University of Tennessee Knoxville, Knoxville, TN, USA
| | - Xuejun Qin
- Durham Veterans Affairs Health Care System, Durham, NC, USA
- Duke University School of Medicine, Duke University, Durham, NC, USA
| | - Niamh Mullins
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Anna Docherty
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Hilary Coon
- Department of Psychiatry, University of Utah School of Medicine, Salt Lake City, UT, USA
- Huntsman Mental Health Institute, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Douglas M Ruderfer
- Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Michael R Garvin
- Computational and Predictive Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
| | - John P Pestian
- Computational and Predictive Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA
- Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Allison E Ashley-Koch
- Duke University School of Medicine, Duke University, Durham, NC, USA
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Jean C Beckham
- Durham Veterans Affairs Health Care System, Durham, NC, USA
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
- VISN 6 Mid-Atlantic Mental Illness Research, Durham Veterans Affairs Health Care System, Durham, NC, USA
| | - Benjamin McMahon
- Theoretical Biology and Biophysics, Los Alamos National Laboratory, Los Alamos, NM, USA
| | - David W Oslin
- VISN 4 Mental Illness Research, Education, and Clinical Center, Center of Excellence, Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nathan A Kimbrel
- Durham Veterans Affairs Health Care System, Durham, NC, USA.
- Duke University School of Medicine, Duke University, Durham, NC, USA.
- VISN 6 Mid-Atlantic Mental Illness Research, Durham Veterans Affairs Health Care System, Durham, NC, USA.
- VA Health Services Research and Development Center of Innovation to Accelerate Discovery and Practice Transformation, Durham, NC, USA.
| | - Daniel A Jacobson
- Computational and Predictive Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA.
| | - David Kainer
- Computational and Predictive Biology, Oak Ridge National Laboratory, Oak Ridge, TN, USA.
- Centre of Excellence for Plant Success in Nature and Agriculture, University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
29
|
Xiao H, Li L, Yang M, Zhang X, Zhou J, Zeng J, Zhou Y, Lan X, Liu J, Lin Y, Zhong Y, Zhang X, Wang L, Cao Z, Liu P, Mei H, Cai M, Cai X, Tao Y, Zhu Y, Yu C, Hu L, Wang Y, Huang Y, Su F, Gao Y, Zhou R, Xu X, Yang H, Wang J, Zhu H, Zhou A, Jin X. Genetic analyses of 104 phenotypes in 20,900 Chinese pregnant women reveal pregnancy-specific discoveries. CELL GENOMICS 2024; 4:100633. [PMID: 39389017 PMCID: PMC11602630 DOI: 10.1016/j.xgen.2024.100633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 12/14/2023] [Accepted: 07/22/2024] [Indexed: 10/12/2024]
Abstract
Monitoring biochemical phenotypes during pregnancy is vital for maternal and fetal health, allowing early detection and management of pregnancy-related conditions to ensure safety for both. Here, we conducted a genetic analysis of 104 pregnancy phenotypes in 20,900 Chinese women. The genome-wide association study (GWAS) identified a total of 410 trait-locus associations, with 71.71% reported previously. Among the 116 novel hits for 45 phenotypes, 83 were successfully replicated. Among them, 31 were defined as potentially pregnancy-specific associations, including creatine and HELLPAR and neutrophils and ESR1, with subsequent analysis revealing enrichments in estrogen-related pathways and female reproductive tissues. The partitioning heritability underscored the significant roles of fetal blood, embryoid bodies, and female reproductive organs in pregnancy hematology and birth outcomes. Pathway analysis confirmed the intricate interplay of hormone and immune regulation, metabolism, and cell cycle during pregnancy. This study contributes to the understanding of genetic influences on pregnancy phenotypes and their implications for maternal health.
Collapse
Affiliation(s)
- Han Xiao
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | - Linxuan Li
- BGI Research, Shenzhen 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Meng Yang
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | - Xinyi Zhang
- BGI Research, Shenzhen 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jieqiong Zhou
- Department of Obstetrics, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | - Jingyu Zeng
- BGI Research, Shenzhen 518083, China; College of Life Sciences, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yan Zhou
- Department of Obstetrics, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | - Xianmei Lan
- BGI Research, Shenzhen 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jiuying Liu
- Department of Obstetrics, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | - Ying Lin
- BGI Research, Shenzhen 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuanyuan Zhong
- Department of Obstetrics, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | - Xiaoqian Zhang
- BGI Research, Shenzhen 518083, China; College of Computer Science and Technology, Guizhou University, Guiyang 550025, China
| | - Lin Wang
- BGI Research, Shenzhen 518083, China
| | - Zhongqiang Cao
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | | | - Hong Mei
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | | | - Xiaonan Cai
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | - Ye Tao
- BGI Research, Shenzhen 518083, China
| | - Yunqing Zhu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing 100191, China
| | - Canqing Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Peking University Health Science Center, Beijing 100191, China; Center for Public Health and Epidemic Preparedness & Response, Peking University, Beijing 100191, China
| | - Liqin Hu
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China
| | - Yu Wang
- BGI Research, Shenzhen 518083, China
| | - Yushan Huang
- BGI Research, Shenzhen 518083, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Ya Gao
- BGI Research, Shenzhen 518083, China
| | | | - Xun Xu
- BGI Research, Shenzhen 518083, China; Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen 518120, China
| | - Huanming Yang
- BGI Research, Shenzhen 518083, China; Guangdong Provincial Academician Workstation of BGI Synthetic Genomics, BGI, Shenzhen 518120, China; James D. Watson Institute of Genome Sciences, Hangzhou 310058, China
| | | | - Huanhuan Zhu
- BGI Research, Shenzhen 518083, China; BGI Research, Wuhan 430074, China.
| | - Aifen Zhou
- Institute of Maternal and Child Health, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China; Department of Obstetrics, Wuhan Children's Hospital (Wuhan Maternal and Child Health Care Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430010, China.
| | - Xin Jin
- BGI Research, Shenzhen 518083, China; BGI Research, Wuhan 430074, China; The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou 510006, China; Shanxi Medical University-BGI Collaborative Center for Future Medicine, Shanxi Medical University, Taiyuan 030001, China; Shenzhen Key Laboratory of Transomics Biotechnologies, BGI Research, Shenzhen 518083, China.
| |
Collapse
|
30
|
Zhang YY, Chen BX, Wan Q. Association of lipid-lowering drugs with the risk of type 2 diabetes and its complications: a mendelian randomized study. Diabetol Metab Syndr 2024; 16:240. [PMID: 39367514 PMCID: PMC11451088 DOI: 10.1186/s13098-024-01477-8] [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: 06/04/2024] [Accepted: 09/23/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND The pathogenesis of type 2 diabetes mellitus is somewhat associated with lipid metabolism. We aim to assess the impact of lipid-lowering drugs (HMGCR inhibitors, PCSK9 inhibitors, and NPC1L1 inhibitors) on type 2 diabetes mellitus and its complications through a two-sample Mendelian randomization (MR) study. METHOD We identified suitable genetic instruments from the GWAS database that represent the expression levels of three genes, interpreting reduced genetically proxied gene expression as indicative of lipid-lowering drug use. We evaluated the causal relationships among these variables employing a two-sample Mendelian randomization approach, with the Inverse Variance Weighted (IVW) analysis serving as the primary method. Coronary artery disease was utilized as a positive control to validate the reliability of the selected genetic instruments. RESULT Increased genetically proxied HMGCR expression is significantly associated with a reduced risk of type 2 diabetes mellitus (OR = 0.64, 95%CI = 0.55-0.74), which was replicated in the FinnGen study with consistent results (OR = 0.65, 95%CI = 0.53-0.80). Increased genetically proxied HMGCR expression is associated with a reduced risk of diabetic retinopathy (OR = 0.23, 95%CI = 0.12-0.44) and diabetic nephropathy (OR = 0.35, 95%CI = 0.17-0.71). In contrast, increased genetically proxied PCSK9 expression is associated with a decreased risk of diabetic coma (OR = 0.70, 95%CI = 0.50-0.98), diabetic neuropathy (OR = 0.24, 95%CI = 0.14-0.42), diabetic retinopathy (OR = 0.67, 95%CI = 0.48-0.96), diabetic cardiovascular diseases (OR = 0.62, 95%CI = 0.38-0.99), and diabetic nephropathy (OR = 0.62, 95%CI = 0.41-0.95). CONCLUSIONS This Mendelian randomization study suggests an association between HMGCR and the pathogenesis of type 2 diabetes mellitus, with increased genetically proxied HMGCR expression reducing the risk of type 2 diabetes mellitus, while PCSK9 and NPC1L1 show no significant association with type 2 diabetes mellitus. These findings may offer more reasonable lipid-lowering drug options for patients with dyslipidemia.
Collapse
Affiliation(s)
- Yue-Yang Zhang
- Department of Endocrinology and Metabolism, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, 646000, China
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, 646000, China
- Sichuan Clinical Research Center for Nephropathy, Luzhou, 646000, China
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, 646000, China
| | - Bing-Xue Chen
- Department of Ultrasound Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Qin Wan
- Department of Endocrinology and Metabolism, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
- Metabolic Vascular Disease Key Laboratory of Sichuan Province, Luzhou, 646000, China.
- Sichuan Clinical Research Center for Diabetes and Metabolism, Luzhou, 646000, China.
- Sichuan Clinical Research Center for Nephropathy, Luzhou, 646000, China.
- Cardiovascular and Metabolic Diseases Key Laboratory of Luzhou, Luzhou, 646000, China.
| |
Collapse
|
31
|
Imamura M, Maeda S. Genetic studies of type 2 diabetes, and microvascular complications of diabetes. Diabetol Int 2024; 15:699-706. [PMID: 39469559 PMCID: PMC11512959 DOI: 10.1007/s13340-024-00727-4] [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: 01/16/2024] [Accepted: 04/24/2024] [Indexed: 10/30/2024]
Abstract
Genome-wide association studies (GWAS) have significantly advanced the identification of genetic susceptibility variants associated with complex diseases. As of 2023, approximately 800 variants predisposing individuals to the risk of type 2 diabetes (T2D) were identified through GWAS, and the majority of studies were predominantly conducted in European populations. Despite the shared nature of the majority of genetic susceptibility loci across diverse ethnic populations, GWAS in non-European populations, including Japanese and East Asian populations, have revealed population-specific T2D loci. Currently, polygenic risk scores (PRSs), encompassing millions of associated variants, can identify individuals with a higher T2D risk than the general population. However, GWAS focusing on microvascular complications of diabetes have identified a limited number of disease-susceptibility loci. Ongoing efforts are crucial to enhance the applicability of PRS for all ethnic groups and unravel the genetic architecture of microvascular complications of diabetes.
Collapse
Affiliation(s)
- Minako Imamura
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of Medicine, University of the Ryukyus, Nishihara-Cho, Okinawa 903-0215 Japan
- Division of Clinical Laboratory and Blood Transfusion, University of the Ryukyus Hospital, Nishihara-Cho, Okinawa 930-0215 Japan
| | - Shiro Maeda
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of Medicine, University of the Ryukyus, Nishihara-Cho, Okinawa 903-0215 Japan
- Division of Clinical Laboratory and Blood Transfusion, University of the Ryukyus Hospital, Nishihara-Cho, Okinawa 930-0215 Japan
| |
Collapse
|
32
|
Wu T, Hu Y, Tang LV. Gene therapy for polygenic or complex diseases. Biomark Res 2024; 12:99. [PMID: 39232780 PMCID: PMC11375922 DOI: 10.1186/s40364-024-00618-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/10/2024] [Indexed: 09/06/2024] Open
Abstract
Gene therapy utilizes nucleic acid drugs to treat diseases, encompassing gene supplementation, gene replacement, gene silencing, and gene editing. It represents a distinct therapeutic approach from traditional medications and introduces novel strategies for genetic disorders. Over the past two decades, significant advancements have been made in the field of gene therapy, leading to the approval of various gene therapy drugs. Gene therapy was initially employed for treating genetic diseases and cancers, particularly monogenic conditions classified as orphan diseases due to their low prevalence rates; however, polygenic or complex diseases exhibit higher incidence rates within populations. Extensive research on the etiology of polygenic diseases has unveiled new therapeutic targets that offer fresh opportunities for their treatment. Building upon the progress achieved in gene therapy for monogenic diseases and cancers, extending its application to polygenic or complex diseases would enable targeting a broader range of patient populations. This review aims to discuss the strategies of gene therapy, methods of gene editing (mainly CRISPR-CAS9), and carriers utilized in gene therapy, and highlight the applications of gene therapy in polygenic or complex diseases focused on applications that have either entered clinical stages or are currently undergoing clinical trials.
Collapse
Affiliation(s)
- Tingting Wu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Key Laboratory of Biological Targeted Therapies of the Chinese Ministry of Education, Wuhan, China
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapies of the Chinese Ministry of Education, Wuhan, China.
| | - Liang V Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Key Laboratory of Biological Targeted Therapies of the Chinese Ministry of Education, Wuhan, China.
| |
Collapse
|
33
|
Guo S, Zhang J, Li H, Cheng CK, Zhang J. Genetic and Modifiable Risk Factors for Postoperative Complications of Total Joint Arthroplasty: A Genome-Wide Association and Mendelian Randomization Study. Bioengineering (Basel) 2024; 11:797. [PMID: 39199755 PMCID: PMC11351150 DOI: 10.3390/bioengineering11080797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024] Open
Abstract
Background: Total joint arthroplasty (TJA) is an orthopedic procedure commonly used to treat damaged joints. Despite the efficacy of TJA, postoperative complications, including aseptic prosthesis loosening and infections, are common. Moreover, the effects of individual genetic susceptibility and modifiable risk factors on these complications are unclear. This study analyzed these effects to enhance patient prognosis and postoperative management. Methods: We conducted an extensive genome-wide association study (GWAS) and Mendelian randomization (MR) study using UK Biobank data. The cohort included 2964 patients with mechanical complications post-TJA, 957 with periprosthetic joint infection (PJI), and a control group of 398,708 individuals. Genetic loci associated with postoperative complications were identified by a GWAS analysis, and the causal relationships of 11 modifiable risk factors with complications were assessed using MR. Results: The GWAS analysis identified nine loci associated with post-TJA complications. Two loci near the PPP1R3B and RBM26 genes were significantly linked to mechanical complications and PJI, respectively. The MR analysis demonstrated that body mass index was positively associated with the risk of mechanical complications (odds ratio [OR]: 1.42; p < 0.001). Higher educational attainment was associated with a decreased risk of mechanical complications (OR: 0.55; p < 0.001) and PJI (OR: 0.43; p = 0.001). Type 2 diabetes was suggestively associated with mechanical complications (OR, 1.18, p = 0.02), and hypertension was suggestively associated with PJI (OR, 1.41, p = 0.008). Other lifestyle factors, including smoking and alcohol consumption, were not causally related to postoperative complications. Conclusions: The genetic loci near PPP1R3B and RBM26 influenced the risk of post-TJA mechanical complications and infections, respectively. The effects of genetic and modifiable risk factors, including body mass index and educational attainment, underscore the need to perform personalized preoperative assessments and the postoperative management of surgical patients. These results indicate that integrating genetic screening and lifestyle interventions into patient care can improve the outcomes of TJA and patient quality of life.
Collapse
Affiliation(s)
- Sijia Guo
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; (S.G.); (J.Z.)
- Engineering Research Center of Digital Medicine of the Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jiping Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; (S.G.); (J.Z.)
- Engineering Research Center of Digital Medicine of the Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Huiwu Li
- Department of Orthopaedics, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China;
| | - Cheng-Kung Cheng
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China; (S.G.); (J.Z.)
- Engineering Research Center of Digital Medicine of the Ministry of Education, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jingwei Zhang
- Department of Orthopaedics, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, 639 Zhizaoju Road, Shanghai 200011, China;
| |
Collapse
|
34
|
Neikirk K, Kabugi K, Mungai M, Kula B, Smith N, Hinton AO. Ethnicity-related differences in mitochondrial regulation by insulin stimulation in diabetes. J Cell Physiol 2024; 239:e31317. [PMID: 38775168 PMCID: PMC11324399 DOI: 10.1002/jcp.31317] [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: 11/23/2023] [Revised: 04/26/2024] [Accepted: 05/07/2024] [Indexed: 08/15/2024]
Abstract
Mitochondrial dysfunction has long been implicated in the development of insulin resistance, which is a hallmark of type 2 diabetes. However, recent studies reveal ethnicity-related differences in mitochondrial processes, underscoring the need for nuance in studying mitochondrial dysfunction and insulin sensitivity. Furthermore, the higher prevalence of type 2 diabetes among African Americans and individuals of African descent has brought attention to the role of ethnicity in disease susceptibility. In this review, which covers existing literature, genetic studies, and clinical data, we aim to elucidate the complex relationship between mitochondrial alterations and insulin stimulation by considering how mitochondrial dynamics, contact sites, pathways, and metabolomics may be differentially regulated across ethnicities, through mechanisms such as single nucleotide polymorphisms (SNPs). In addition to achieving a better understanding of insulin stimulation, future studies identifying novel regulators of mitochondrial structure and function could provide valuable insights into ethnicity-dependent insulin signaling and personalized care.
Collapse
Affiliation(s)
- Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Kinuthia Kabugi
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Margaret Mungai
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Bartosz Kula
- Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester, School of Medicine and Dentistry, Rochester, USA 14642
| | - Nathan Smith
- Del Monte Institute for Neuroscience, Department of Neuroscience, University of Rochester, School of Medicine and Dentistry, Rochester, USA 14642
| | - Antentor O. Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| |
Collapse
|
35
|
Marigorta UM, Millet O, Lu SC, Mato JM. Dysfunctional VLDL metabolism in MASLD. NPJ METABOLIC HEALTH AND DISEASE 2024; 2:16. [PMID: 39049993 PMCID: PMC11263124 DOI: 10.1038/s44324-024-00018-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 06/22/2024] [Indexed: 07/27/2024]
Abstract
Lipidomics has unveiled the intricate human lipidome, emphasizing the extensive diversity within lipid classes in mammalian tissues critical for cellular functions. This diversity poses a challenge in maintaining a delicate balance between adaptability to recurring physiological changes and overall stability. Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD), linked to factors such as obesity and diabetes, stems from a compromise in the structural and functional stability of the liver within the complexities of lipid metabolism. This compromise inaccurately senses an increase in energy status, such as during fasting-feeding cycles or an upsurge in lipogenesis. Serum lipidomic studies have delineated three distinct metabolic phenotypes, or "metabotypes" in MASLD. MASLD-A is characterized by lower very low-density lipoprotein (VLDL) secretion and triglyceride (TG) levels, associated with a reduced risk of cardiovascular disease (CVD). In contrast, MASLD-C exhibits increased VLDL secretion and TG levels, correlating with elevated CVD risk. An intermediate subtype, with a blend of features, is designated as the MASLD-B metabotype. In this perspective, we examine into recent findings that show the multifaceted regulation of VLDL secretion by S-adenosylmethionine, the primary cellular methyl donor. Furthermore, we explore the differential CVD and hepatic cancer risk across MASLD metabotypes and discuss the context and potential paths forward to gear the findings from genetic studies towards a better understanding of the observed heterogeneity in MASLD.
Collapse
Affiliation(s)
- Urko M. Marigorta
- Integrative Genomics Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), 48160 Derio, Spain
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
| | - Oscar Millet
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), CIBERehd, 48160 Derio, Spain
| | - Shelly C. Lu
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA 90048 USA
| | - José M. Mato
- Precision Medicine and Metabolism Lab, CIC bioGUNE, Basque Research and Technology Alliance (BRTA), CIBERehd, 48160 Derio, Spain
| |
Collapse
|
36
|
Baradaran Mahdavi S, Javadirad SM, Rafieian M, Poursafa P, Azimian Zavareh V, Daniali SS, Heidari-Beni M, Goodarzi-Khoigani M, Vahdatpour B, Mirhendi H, Kelishadi R. A procedure for DNA methylation assessment in osteoporosis-related gene promoters of umbilical cord blood: A study on the Prospective Epidemiological Research Studies in Iran (PERSIAN) birth cohort. BIOIMPACTS : BI 2024; 15:30095. [PMID: 40161946 PMCID: PMC11954747 DOI: 10.34172/bi.30095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/10/2023] [Accepted: 01/02/2024] [Indexed: 04/02/2025]
Abstract
Introduction It is believed that DNA methylation can modify disease susceptibility in response to environmental factors as early as the perinatal period. In this study, we aimed to present a streamlined DNA methylation analysis procedure for osteoporosis-related gene promoters in the umbilical cord blood. Methods The Prospective Epidemiological Research Studies in Iran (PERSIAN) birth cohort was established in 2016. In this study, a total of 300 umbilical cord blood samples were collected at the time of delivery. For all samples, DNA was extracted and converted using sodium bisulfite. Multiple primer sets were designed for Wnt1, Wnt10b, β-catenin, OPG, and RANKL gene promoters in the online MethPrimer platform. Next, bisulfite sequencing PCR (BSP), as the gold standard method for exploring methylated and unmethylated cytosines, was performed in a gradient-controlled setting. The PCR products were then purified and directly sequenced. Subsequently, the chromatograms were interpreted. Results For Wnt10b, β-catenin, and OPG genes, the converted DNA could be successfully amplified. The frequency of acceptable chromatograms for analysis was 195 for Wnt10b (195/300, 0.65%), 198 for β-catenin (198/300, 0.66%), and 50 for OPG (50/50, 100%). Conclusion BSP can be efficiently used to investigate the methylation of target gene promoters in umbilical cord blood DNA.
Collapse
Affiliation(s)
- Sadegh Baradaran Mahdavi
- Department of Physical Medicine and Rehabilitation, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
- Core Research Facilities (CRF), Isfahan University of Medical Sciences, Isfahan, Iran
| | - Seyed Morteza Javadirad
- Department of Cell and Molecular Biology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Mahsa Rafieian
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
- Core Research Facilities (CRF), Isfahan University of Medical Sciences, Isfahan, Iran
| | - Parnian Poursafa
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Seyede Shahrbanoo Daniali
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Motahar Heidari-Beni
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Masoomeh Goodarzi-Khoigani
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Babak Vahdatpour
- Department of Physical Medicine and Rehabilitation, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Mirhendi
- Core Research Facilities (CRF), Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Medical Parasitology and Mycology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Roya Kelishadi
- Child Growth and Development Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
37
|
Wen R, Xi YJ, Zhang R, Hou SJ, Shi JY, Chen JY, Zhang HY, Qiao J, Feng YQ, Zhang SX. Prescription glucocorticoid medication and iridocyclitis are associated with an increased risk of senile cataract occurrence: a Mendelian randomization study. Aging (Albany NY) 2024; 16:10563-10578. [PMID: 38925660 PMCID: PMC11236313 DOI: 10.18632/aging.205963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 05/06/2024] [Indexed: 06/28/2024]
Abstract
Iridocyclitis and the use of glucocorticoid medication have been widely studied as susceptibility factors for cataracts. However, the causal relationship between them remains unclear. This study aimed to investigate the causal relationship between the development of iridocyclitis and the genetic liability of glucocorticoid medication use on the risk of senile cataracts occurrence by performing Two-sample Mendelian randomization (MR) analyses. Instrumental variables (IVs) significantly associated with exposure factors (P < 5 × 10-8) were identified using published genome-wide association data from the FinnGen database and UK Biobank. Reliability analyses were conducted using five approaches, including inverse-variance weighted (IVW), MR-Egger regression, simple median, weighted median, and weighted mode. A sensitivity analysis using the leave-one-out method was also performed. Genetic susceptibility to glucocorticoid use was associated with an increased risk of developing senile cataracts (OR, 1.10; 95% CI, 1.02-1.17; P < 0.05). Moreover, iridocyclitis was significantly associated with a higher risk of developing senile cataracts (OR, 1.03; 95% CI, 1.01-1.05; P < 0.05). Nonetheless, some heterogeneity in the IVs was observed, but the MR results remained consistent after penalizing for outliers. The estimates were consistent in multivariate analyses by adjusting for body mass index (BMI) and diabetes mellitus type 2 (T2DM). This study provides new insights into the prevention and management of senile cataracts by highlighting the increased risk associated with iridocyclitis and the use of glucocorticoids.
Collapse
Affiliation(s)
- Rui Wen
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi, China
| | - Yu-Jia Xi
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi, China
| | - Ran Zhang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi, China
| | - Si-Jia Hou
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi, China
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jin-Yu Shi
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi, China
- Department of Breast Surgery, Fifth Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jin-Yi Chen
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi, China
| | - He-Yi Zhang
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi, China
| | - Jun Qiao
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi, China
| | - Yi-Qian Feng
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi, China
| | - Sheng-Xiao Zhang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
- Shanxi Provincial Key Laboratory of Rheumatism Immune Microecology, Taiyuan, Shanxi, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi, China
| |
Collapse
|
38
|
Li F, Wang Y, Cao J, Chen Q, Gao Y, Li R, Yuan L. Integrated analysis of genes shared between type 2 diabetes mellitus and osteoporosis. Front Pharmacol 2024; 15:1388205. [PMID: 38966541 PMCID: PMC11222565 DOI: 10.3389/fphar.2024.1388205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/16/2024] [Indexed: 07/06/2024] Open
Abstract
Background The relationship between type 2 diabetes mellitus (T2DM) and osteoporosis (OP) has been widely recognized in recent years, but the mechanism of interaction remains unknown. The aim of this study was to investigate the genetic features and signaling pathways that are shared between T2DM and OP. Methods We analyzed the GSE76894 and GSE76895 datasets for T2DM and GSE56815 and GSE7429 for OP from the Gene Expression Omnibus (GEO) database to identify shared genes in T2DM and OP, and we constructed coexpression networks based on weighted gene coexpression network analysis (WGCNA). Shared genes were then further analyzed for functional pathway enrichment. We selected the best common biomarkers using the least absolute shrinkage and selection operator (LASSO) algorithm and validated the common biomarkers, followed by RT-PCR, immunofluorescence, Western blotting, and enzyme-linked immunosorbent assay (ELISA) to validate the expression of these hub genes in T2DM and OP mouse models and patients. Results We found 8,506 and 2,030 DEGs in T2DM and OP, respectively. Four modules were identified as significant for T2DM and OP using WGCNA. A total of 19 genes overlapped with the strongest positive and negative modules of T2DM and OP. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed these genes may be involved in pantothenate and CoA biosynthesis and the glycosaminoglycan biosynthesis-chondroitin sulfate/dermatan sulfate and renin-angiotensin system signaling pathway. The LASSO algorithm calculates the six optimal common biomarkers. RT-PCR results show that LTB, TPBG, and VNN1 were upregulated in T2DM and OP. Immunofluorescence and Western blot show that VNN1 is upregulated in the pancreas and bones of T2DM model mice and osteoporosis model mice. Similarly, the level of VNN1 in the sera of patients with T2DM, OP, and T2DM and OP was higher than that in the healthy group. Conclusion Based on the WGCNA and LASSO algorithms, we identified genes and pathways that were shared between T2DM and OP. Both pantothenate and CoA biosynthesis and the glycosaminoglycan biosynthesis-chondroitin sulfate/dermatan sulfate and renin-angiotensin systems may be associated with the pathogenesis of T2DM and OP. Moreover, VNN1 may be a potential diagnostic marker for patients with T2DM complicated by OP. This study provides a new perspective for the systematic study of possible mechanisms of combined OP and T2DM.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Li Yuan
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
39
|
Annicchiarico A, Barile B, Buccoliero C, Nicchia GP, Brunetti G. Alternative therapeutic strategies in diabetes management. World J Diabetes 2024; 15:1142-1161. [PMID: 38983831 PMCID: PMC11229975 DOI: 10.4239/wjd.v15.i6.1142] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 02/17/2024] [Accepted: 04/12/2024] [Indexed: 06/11/2024] Open
Abstract
Diabetes is a heterogeneous metabolic disease characterized by elevated blood glucose levels resulting from the destruction or malfunction of pancreatic β cells, insulin resistance in peripheral tissues, or both, and results in a non-sufficient production of insulin. To adjust blood glucose levels, diabetic patients need exogenous insulin administration together with medical nutrition therapy and physical activity. With the aim of improving insulin availability in diabetic patients as well as ameliorating diabetes comorbidities, different strategies have been investigated. The first approaches included enhancing endogenous β cell activity or transplanting new islets. The protocol for this kind of intervention has recently been optimized, leading to standardized procedures. It is indicated for diabetic patients with severe hypoglycemia, complicated by impaired hypoglycemia awareness or exacerbated glycemic lability. Transplantation has been associated with improvement in all comorbidities associated with diabetes, quality of life, and survival. However, different trials are ongoing to further improve the beneficial effects of transplantation. Furthermore, to overcome some limitations associated with the availability of islets/pancreas, alternative therapeutic strategies are under evaluation, such as the use of mesenchymal stem cells (MSCs) or induced pluripotent stem cells for transplantation. The cotransplantation of MSCs with islets has been successful, thus providing protection against proinflammatory cytokines and hypoxia through different mechanisms, including exosome release. The use of induced pluripotent stem cells is recent and requires further investigation. The advantages of MSC implantation have also included the improvement of diabetes-related comorbidities, such as wound healing. Despite the number of advantages of the direct injection of MSCs, new strategies involving biomaterials and scaffolds have been developed to improve the efficacy of mesenchymal cell delivery with promising results. In conclusion, this paper offered an overview of new alternative strategies for diabetes management while highlighting some limitations that will need to be overcome by future approaches.
Collapse
Affiliation(s)
- Alessia Annicchiarico
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Bari 70125, Italy
| | - Barbara Barile
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Bari 70125, Italy
| | - Cinzia Buccoliero
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Bari 70125, Italy
| | - Grazia Paola Nicchia
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Bari 70125, Italy
| | - Giacomina Brunetti
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, Bari 70125, Italy
| |
Collapse
|
40
|
Aliyu U, Umlai UKI, Toor SM, Elashi AA, Al-Sarraj YA, Abou−Samra AB, Suhre K, Albagha OME. Genome-wide association study and polygenic score assessment of insulin resistance. Front Endocrinol (Lausanne) 2024; 15:1384103. [PMID: 38938516 PMCID: PMC11208314 DOI: 10.3389/fendo.2024.1384103] [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: 03/15/2024] [Accepted: 05/14/2024] [Indexed: 06/29/2024] Open
Abstract
Insulin resistance (IR) and beta cell dysfunction are the major drivers of type 2 diabetes (T2D). Genome-Wide Association Studies (GWAS) on IR have been predominantly conducted in European populations, while Middle Eastern populations remain largely underrepresented. We conducted a GWAS on the indices of IR (HOMA2-IR) and beta cell function (HOMA2-%B) in 6,217 non-diabetic individuals from the Qatar Biobank (QBB; Discovery cohort; n = 2170, Replication cohort; n = 4047) with and without body mass index (BMI) adjustment. We also developed polygenic scores (PGS) for HOMA2-IR and compared their performance with a previously derived PGS for HOMA-IR (PGS003470). We replicated 11 loci that have been previously associated with HOMA-IR and 24 loci that have been associated with HOMA-%B, at nominal statistical significance. We also identified a novel locus associated with beta cell function near VEGFC gene, tagged by rs61552983 (P = 4.38 × 10-8). Moreover, our best performing PGS (Q-PGS4; Adj R2 = 0.233 ± 0.014; P = 1.55 x 10-3) performed better than PGS003470 (Adj R2 = 0.194 ± 0.014; P = 5.45 x 10-2) in predicting HOMA2-IR in our dataset. This is the first GWAS on HOMA2 and the first GWAS conducted in the Middle East focusing on IR and beta cell function. Herein, we report a novel locus in VEGFC that is implicated in beta cell dysfunction. Inclusion of under-represented populations in GWAS has potentials to provide important insights into the genetic architecture of IR and beta cell function.
Collapse
Affiliation(s)
- Usama Aliyu
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Umm-Kulthum Ismail Umlai
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Salman M. Toor
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Asma A. Elashi
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| | - Yasser A. Al-Sarraj
- Qatar Genome Program (QGP), Qatar Foundation Research, Development and Innovation, Qatar Foundation (QF), Doha, Qatar
| | | | - Karsten Suhre
- Bioinformatics Core, Weill Cornell Medicine-Qatar, Doha, Qatar
- Department of Biophysics and Physiology, Weill Cornell Medicine, New York, NY, United States
| | - Omar M. E. Albagha
- College of Health and Life Sciences (CHLS), Hamad Bin Khalifa University (HBKU), Qatar Foundation (QF), Doha, Qatar
| |
Collapse
|
41
|
Sun W, Wang Y, Li C, Yao X, Wu X, He A, Zhao B, Huang X, Song H. Genetically predicted high serum sex hormone-binding globulin levels are associated with lower ischemic stroke risk: A sex-stratified Mendelian randomization study. J Stroke Cerebrovasc Dis 2024; 33:107686. [PMID: 38522757 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 03/17/2024] [Accepted: 03/19/2024] [Indexed: 03/26/2024] Open
Abstract
OBJECTIVE Cross-sectional and cohort studies have found insufficient evidence of a causal relationship between sex hormone-binding globulin and ischemic stroke, only associations. Here, we performed a sex-stratified, bidirectional, two-sample Mendelian randomization analysis to evaluate whether a causal relationship exists between sex hormone-binding globulin and ischemic stroke. METHODS Single-nucleotide polymorphisms associated with sex hormone-binding globulin and ischemic stroke were screened from genome-wide association studies summary data as instrumental variables to enable a bidirectional, two-sample Mendelian randomization study design. Inverse-variance weighted analysis was used as the main method to evaluate potential causality, and additional methods, including the weighted median and MR-Egger tests, were used to validate the Mendelian randomization results. Cochran's Q statistic, MR-Egger intercept test, and Mendelian Randomization-Pleiotropy Residual Sum and Outlier global test were used as sensitivity analysis techniques to assure the reliability of the results. Multivariable analysis was used to show the robustness of the results with key theorized confounders. RESULTS Inverse-variance weighted analysis showed that genetically predicted higher serum sex hormone-binding globulin levels were associated with significantly decreased risk of ischemic stroke in males (odds radio = 0.934, 95 % confidence interval = 0.885-0.985, P = 0.012) and females (odds radio = 0.924, 95 % confidence interval = 0.868-0.983, P = 0.013). In an analysis of ischemic stroke subtypes, genetically predicted higher serum sex hormone-binding globulin levels were also associated with significantly decreased risk of small-vessel occlusion in both males (odds radio = 0.849, 95 % confidence interval = 0.759-0.949, P = 0.004) and females (odds radio = 0.829, 95 % confidence interval = 0.724-0.949, P = 0.006). The association remained in sensitivity analyses and multivariable analyses. The reverse analysis suggested an association between genetically predicted risk of cardioembolism and increased serum sex hormone-binding globulin in females (Beta = 0.029 nmol/L, Standard Error = 0.010, P = 0.003). CONCLUSION Our findings provide new insight into the etiology of ischemic stroke and suggest that modulating serum sex hormone-binding globulin may be a therapeutic strategy to protect against ischemic stroke.
Collapse
Affiliation(s)
- Wei Sun
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Yuan Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Cancan Li
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, Beijing 100069, China
| | - Xuefan Yao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xiao Wu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Aini He
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Benke Zhao
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Xiaoqin Huang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China
| | - Haiqing Song
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| |
Collapse
|
42
|
Kui L, Dong C, Wu J, Zhuo F, Yan B, Wang Z, Yang M, Xiong C, Qiu P. Causal association between type 2 diabetes mellitus and acute suppurative otitis media: insights from a univariate and multivariate Mendelian randomization study. Front Endocrinol (Lausanne) 2024; 15:1407503. [PMID: 38836234 PMCID: PMC11148255 DOI: 10.3389/fendo.2024.1407503] [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: 03/26/2024] [Accepted: 05/09/2024] [Indexed: 06/06/2024] Open
Abstract
Background Type 2 diabetes mellitus (T2DM) and hearing loss (HL) constitute significant public health challenges worldwide. Recently, the association between T2DM and HL has aroused attention. However, possible residual confounding factors and other biases inherent to observational study designs make this association undetermined. In this study, we performed univariate and multivariable Mendelian Randomization (MR) analysis to elucidate the causal association between T2DM and common hearing disorders that lead to HL. Methods Our study employed univariate and multivariable MR analyses, with the Inverse Variance Weighted method as the primary approach to assessing the potential causal association between T2DM and hearing disorders. We selected 164 and 9 genetic variants representing T2DM from the NHGRI-EBI and DIAGRAM consortium, respectively. Summary-level data for 10 hearing disorders were obtained from over 500,000 participants in the FinnGen consortium and MRC-IEU. Sensitivity analysis revealed no significant heterogeneity of instrumental variables or pleiotropy was detected. Results In univariate MR analysis, genetically predicted T2DM from both sources was associated with an increased risk of acute suppurative otitis media (ASOM) (In NHGRI-EBI: OR = 1.07, 95% CI: 1.02-1.13, P = 0.012; In DIAGRAM: OR = 1.14, 95% CI: 1.02-1.26, P = 0.016). Multivariable MR analysis, adjusting for genetically predicted sleep duration, alcohol consumption, body mass index, and smoking, either individually or collectively, maintained these associations. Sensitivity analyses confirmed the robustness of the results. Conclusion T2DM was associated with an increased risk of ASOM. Strict glycemic control is essential for the minimization of the effects of T2DM on ASOM.
Collapse
Affiliation(s)
- Lihong Kui
- Xiamen Rehabilitation Hospital, Xiamen, Fujian, China
| | - Cheng Dong
- Depart of Rehabilitation Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junyu Wu
- School of Physical Education, Shanghai University of Sport, Shanghai, China
| | - Feinan Zhuo
- Department of Rehabilitation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Bin Yan
- School of Rehabilitation Medicine, Jiangsu Vocational College of Medicine, Jiangsu, China
| | - Zhewei Wang
- Department of Rehabilitation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Meiling Yang
- Xiamen Rehabilitation Hospital, Xiamen, Fujian, China
| | - Canhai Xiong
- Xiamen Rehabilitation Hospital, Xiamen, Fujian, China
| | - Peng Qiu
- Department of Rehabilitation, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
43
|
Zhang W, Sun J, Yu H, Shi M, Hu H, Yuan H. Causal relationship between type 2 diabetes mellitus and aortic dissection: insights from two-sample Mendelian randomization and mediation analysis. Front Endocrinol (Lausanne) 2024; 15:1405517. [PMID: 38803481 PMCID: PMC11128602 DOI: 10.3389/fendo.2024.1405517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 04/29/2024] [Indexed: 05/29/2024] Open
Abstract
Objective Some evidence suggests a reduced prevalence of type 2 diabetes mellitus (T2DM) in patients with aortic dissection (AD), a catastrophic cardiovascular illness, compared to general population. However, the conclusions were inconsistent, and the causal relationship between T2DM and AD remains unclear. Methods In this study, we aimed to explore the causal relationship between T2DM and AD using bidirectional Mendelian randomization (MR) analysis. Mediation MR analysis was conducted to explore and quantify the possible mediation effects of 1400 metabolites in T2DM and AD. Results The results of 26 datasets showed no causal relationship between T2DM and AD (P>0.05). Only one dataset (ebi-a-GCST90006934) showed that T2DM was a protective factor for AD (I9-AORTDIS) (OR=0.815, 95%CI: 0.692-0.960, P=0.014), and did not show horizontal pleiotropy (P=0.808) and heterogeneity (P=0.525). Vanillic acid glycine plays a mediator in the causal relationship between T2DM and AD. The mediator effect for vanillic acid glycine levels was -0.023 (95%CI: -0.066-0.021). Conclusion From the perspective of MR analysis, there might not be a causal relationship between T2DM and AD, and T2DM might not be a protective factor for AD. If a causal relationship does exist between T2DM and AD, with T2DM serving as a protective factor, vanillic acid glycine may act as a mediator and enhance such a protective effect.
Collapse
Affiliation(s)
| | | | | | | | | | - Hong Yuan
- Department of Cardiovascular, First People’s Hospital of LinPing District, Hangzhou, China
| |
Collapse
|
44
|
Wang SH, Huang YC, Cheng CW, Chang YW, Liao WL. Impact of the trans-ancestry polygenic risk score on type 2 diabetes risk, onset age and progression among population in Taiwan. Am J Physiol Endocrinol Metab 2024; 326:E547-E554. [PMID: 38363735 PMCID: PMC11376485 DOI: 10.1152/ajpendo.00252.2023] [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: 08/14/2023] [Revised: 01/03/2024] [Accepted: 01/05/2024] [Indexed: 02/18/2024]
Abstract
Type 2 diabetes (T2D) prevalence in adults at a younger age has increased but the disease status may go unnoticed. This study aimed to determine whether the onset age and subsequent diabetic complications can be attributed to the polygenic architecture of T2D in the Taiwan Han population. A total of 9,627 cases with T2D and 85,606 controls from the Taiwan Biobank were enrolled. Three diabetic polygenic risk scores (PRSs), PRS_EAS and PRS_EUR, and a trans-ancestry PRS (PRS_META), calculated using summary statistic from East Asian and European populations. The onset age was identified by linking to the National Taiwan Insurance Research Database, and the incidence of different diabetic complications during follow-up was recorded. PRS_META (7.4%) explained a higher variation for T2D status. And the higher percentile of PRS is also correlated with higher percentage of T2D family history and prediabetes status. More, the PRS was negatively associated with onset age (β = -0.91 yr), and this was more evident among males (β = -1.11 vs. -0.76 for males and females, respectively). The hazard ratio of diabetic retinopathy (DR) and diabetic foot were significantly associated with PRS_EAS and PRS_META, respectively. However, the PRS was not associated with other diabetic complications, including diabetic nephropathy, cardiovascular disease, and hypertension. Our findings indicated that diabetic PRS which combined susceptibility variants from cross-population could be used as a tool for early screening of T2D, especially for high-risk populations, such as individuals with high genetic risk, and may be associated with the risk of complications in subjects with T2D. NEW & NOTEWORTHY Our findings indicated that diabetic polygenic risk score (PRS) which combined susceptibility variants from Asian and European population affect the onset age of type 2 diabetes (T2D) and could be used as a tool for early screening of T2D, especially for individuals with high genetic risk, and may be associated with the risk of diabetic complications among people in Taiwan.
Collapse
Affiliation(s)
- Shi-Heng Wang
- National Center for Geriatrics and Welfare Research, National Health Research Institutes, Zhunan, Taiwan
- Department of Public Health, China Medical University, Taichung, Taiwan
| | - Yu-Chuen Huang
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Chun-Wen Cheng
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ya-Wen Chang
- Genetic Center, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Center for Personalized Medicine, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Wen-Ling Liao
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Center for Personalized Medicine, Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
45
|
Bass AJ, Bian S, Wingo AP, Wingo TS, Cutler DJ, Epstein MP. Identifying latent genetic interactions in genome-wide association studies using multiple traits. Genome Med 2024; 16:62. [PMID: 38664839 PMCID: PMC11044415 DOI: 10.1186/s13073-024-01329-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 04/02/2024] [Indexed: 04/28/2024] Open
Abstract
The "missing" heritability of complex traits may be partly explained by genetic variants interacting with other genes or environments that are difficult to specify, observe, and detect. We propose a new kernel-based method called Latent Interaction Testing (LIT) to screen for genetic interactions that leverages pleiotropy from multiple related traits without requiring the interacting variable to be specified or observed. Using simulated data, we demonstrate that LIT increases power to detect latent genetic interactions compared to univariate methods. We then apply LIT to obesity-related traits in the UK Biobank and detect variants with interactive effects near known obesity-related genes (URL: https://CRAN.R-project.org/package=lit ).
Collapse
Affiliation(s)
- Andrew J Bass
- Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA.
| | - Shijia Bian
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, 30322, USA
| | - Aliza P Wingo
- Department of Psychiatry, Emory University, Atlanta, GA, 30322, USA
| | - Thomas S Wingo
- Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA
- Department of Neurology, Emory University, Atlanta, GA, 30322, USA
| | - David J Cutler
- Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA
| | - Michael P Epstein
- Department of Human Genetics, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
46
|
Ke TM, Lophatananon A, Muir KR. Strengthening the Evidence for a Causal Link between Type 2 Diabetes Mellitus and Pancreatic Cancer: Insights from Two-Sample and Multivariable Mendelian Randomization. Int J Mol Sci 2024; 25:4615. [PMID: 38731833 PMCID: PMC11082974 DOI: 10.3390/ijms25094615] [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: 03/20/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 05/13/2024] Open
Abstract
This two-sample Mendelian randomization (MR) study was conducted to investigate the causal associations between type 2 diabetes mellitus (T2DM) and the risk of pancreatic cancer (PaCa), as this causal relationship remains inconclusive in existing MR studies. The selection of instrumental variables for T2DM was based on two genome-wide association study (GWAS) meta-analyses from European cohorts. Summary-level data for PaCa were extracted from the FinnGen and UK Biobank databases. Inverse variance weighted (IVW) and four other robust methods were employed in our MR analysis. Various sensitivity analyses and multivariable MR approaches were also performed to enhance the robustness of our findings. In the IVW and Mendelian Randomization Pleiotropy RESidual Sum and Outlier (MR-PRESSO) analyses, the odds ratios (ORs) for each 1-unit increase in genetically predicted log odds of T2DM were approximately 1.13 for PaCa. The sensitivity tests and multivariable MR supported the causal link between T2DM and PaCa without pleiotropic effects. Therefore, our analyses suggest a causal relationship between T2DM and PaCa, shedding light on the potential pathophysiological mechanisms of T2DM's impact on PaCa. This finding underscores the importance of T2DM prevention as a strategy to reduce the risk of PaCa.
Collapse
Affiliation(s)
| | | | - Kenneth R. Muir
- Division of Population Health, Health Services Research and Primary Care, School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester M13 9PT, UK; (T.-M.K.); (A.L.)
| |
Collapse
|
47
|
Jeong R, Bulyk ML. Chromatin accessibility variation provides insights into missing regulation underlying immune-mediated diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.12.589213. [PMID: 38659802 PMCID: PMC11042205 DOI: 10.1101/2024.04.12.589213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Most genetic loci associated with complex traits and diseases through genome-wide association studies (GWAS) are noncoding, suggesting that the causal variants likely have gene regulatory effects. However, only a small number of loci have been linked to expression quantitative trait loci (eQTLs) detected currently. To better understand the potential reasons for many trait-associated loci lacking eQTL colocalization, we investigated whether chromatin accessibility QTLs (caQTLs) in lymphoblastoid cell lines (LCLs) explain immune-mediated disease associations that eQTLs in LCLs did not. The power to detect caQTLs was greater than that of eQTLs and was less affected by the distance from the transcription start site of the associated gene. Meta-analyzing LCL eQTL data to increase the sample size to over a thousand led to additional loci with eQTL colocalization, demonstrating that insufficient statistical power is still likely to be a factor. Moreover, further eQTL colocalization loci were uncovered by surveying eQTLs of other immune cell types. Altogether, insufficient power and context-specificity of eQTLs both contribute to the 'missing regulation.'
Collapse
Affiliation(s)
- Raehoon Jeong
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Bioinformatics and Integrative Genomics Graduate Program, Harvard University, Cambridge, MA 02138, USA
| | - Martha L. Bulyk
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
- Bioinformatics and Integrative Genomics Graduate Program, Harvard University, Cambridge, MA 02138, USA
- Department of Pathology, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
48
|
Zhao P, Sheng Z, Xu L, Li P, Xiao W, Yuan C, Xu Z, Yang M, Qian Y, Zhong J, Gu J, Karasik D, Zheng HF. Deciphering the complex relationship between type 2 diabetes and fracture risk with both genetic and observational evidence. eLife 2024; 12:RP89281. [PMID: 38591545 PMCID: PMC11003741 DOI: 10.7554/elife.89281] [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] [Indexed: 04/10/2024] Open
Abstract
The 'diabetic bone paradox' suggested that type 2 diabetes (T2D) patients would have higher areal bone mineral density (BMD) but higher fracture risk than individuals without T2D. In this study, we found that the genetically predicted T2D was associated with higher BMD and lower risk of fracture in both weighted genetic risk score (wGRS) and two-sample Mendelian randomization (MR) analyses. We also identified ten genomic loci shared between T2D and fracture, with the top signal at SNP rs4580892 in the intron of gene RSPO3. And the higher expression in adipose subcutaneous and higher protein level in plasma of RSPO3 were associated with increased risk of T2D, but decreased risk of fracture. In the prospective study, T2D was observed to be associated with higher risk of fracture, but BMI mediated 30.2% of the protective effect. However, when stratified by the T2D-related risk factors for fracture, we observed that the effect of T2D on the risk of fracture decreased when the number of T2D-related risk factors decreased, and the association became non-significant if the T2D patients carried none of the risk factors. In conclusion, the genetically determined T2D might not be associated with higher risk of fracture. And the shared genetic architecture between T2D and fracture suggested a top signal around RSPO3 gene. The observed effect size of T2D on fracture risk decreased if the T2D-related risk factors could be eliminated. Therefore, it is important to manage the complications of T2D to prevent the risk of fracture.
Collapse
Affiliation(s)
- Pianpian Zhao
- The affiliated Hangzhou first people’s hospital, School of Medicine, Westlake UniversityHangzhouChina
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, ChinaHangzhouChina
- Westlake Laboratory of Life Sciences and BiomedicineHangzhouChina
- Institute of Basic Medical Sciences, Westlake Institute for Advanced StudyHangzhouChina
| | - Zhifeng Sheng
- Health Management Center, The Second Xiangya Hospital of Central South UniversityChangshaChina
| | - Lin Xu
- Department of Orthopedics, Yantai Affiliated Hospital of Binzhou Medical UniversityYantaiChina
| | - Peng Li
- Department of Geratology, The Third People's Hospital of HangzhouHangzhouChina
| | - Wenjin Xiao
- Department of Endocrinology, Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Chengda Yuan
- Department of Dermatology, Hangzhou Hospital of Traditional Chinese MedicineHangzhouChina
| | - Zhanwei Xu
- Central Health Center of Mashenqiao TownTianjinChina
| | - Mengyuan Yang
- The affiliated Hangzhou first people’s hospital, School of Medicine, Westlake UniversityHangzhouChina
- Westlake Laboratory of Life Sciences and BiomedicineHangzhouChina
- Institute of Basic Medical Sciences, Westlake Institute for Advanced StudyHangzhouChina
| | - Yu Qian
- The affiliated Hangzhou first people’s hospital, School of Medicine, Westlake UniversityHangzhouChina
- Westlake Laboratory of Life Sciences and BiomedicineHangzhouChina
- Institute of Basic Medical Sciences, Westlake Institute for Advanced StudyHangzhouChina
| | - Jiadong Zhong
- The affiliated Hangzhou first people’s hospital, School of Medicine, Westlake UniversityHangzhouChina
- Westlake Laboratory of Life Sciences and BiomedicineHangzhouChina
- Institute of Basic Medical Sciences, Westlake Institute for Advanced StudyHangzhouChina
| | - Jiaxuan Gu
- The affiliated Hangzhou first people’s hospital, School of Medicine, Westlake UniversityHangzhouChina
- Westlake Laboratory of Life Sciences and BiomedicineHangzhouChina
- Institute of Basic Medical Sciences, Westlake Institute for Advanced StudyHangzhouChina
| | - David Karasik
- Azrieli Faculty of Medicine, Bar-Ilan UniversitySafedIsrael
| | - Hou-Feng Zheng
- The affiliated Hangzhou first people’s hospital, School of Medicine, Westlake UniversityHangzhouChina
- Diseases & Population (DaP) Geninfo Lab, School of Life Sciences, Westlake University, Hangzhou, ChinaHangzhouChina
- Westlake Laboratory of Life Sciences and BiomedicineHangzhouChina
- Institute of Basic Medical Sciences, Westlake Institute for Advanced StudyHangzhouChina
| |
Collapse
|
49
|
Imamura M, Maeda S. Perspectives on genetic studies of type 2 diabetes from the genome-wide association studies era to precision medicine. J Diabetes Investig 2024; 15:410-422. [PMID: 38259175 PMCID: PMC10981147 DOI: 10.1111/jdi.14149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/24/2023] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Genome-wide association studies (GWAS) have facilitated a substantial and rapid increase in the number of confirmed genetic susceptibility variants for complex diseases. Approximately 700 variants predisposing individuals to the risk for type 2 diabetes have been identified through GWAS until 2023. From 2018 to 2022, hundreds of type 2 diabetes susceptibility loci with smaller effect sizes were identified through large-scale GWAS with sample sizes of 200,000 to >1 million. The clinical translation of genetic information for type 2 diabetes includes the development of novel therapeutics and risk predictions. Although drug discovery based on loci identified in GWAS remains challenging owing to the difficulty of functional annotation, global efforts have been made to identify novel biological mechanisms and therapeutic targets by applying multi-omics approaches or searching for disease-associated coding variants in isolated founder populations. Polygenic risk scores (PRSs), comprising up to millions of associated variants, can identify individuals with higher disease risk than those in the general population. In populations of European descent, PRSs constructed from base GWAS data with a sample size of approximately 450,000 have predicted the onset of diseases well. However, European GWAS-derived PRSs have limited predictive performance in non-European populations. The predictive accuracy of a PRS largely depends on the sample size of the base GWAS data. The results of GWAS meta-analyses for multi-ethnic groups as base GWAS data and cross-population polygenic prediction methodology have been applied to establish a universal PRS applicable to small isolated ethnic populations.
Collapse
Affiliation(s)
- Minako Imamura
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of MedicineUniversity of the RyukyusNishihara‐ChoJapan
- Division of Clinical Laboratory and Blood TransfusionUniversity of the Ryukyus HospitalNishihara‐ChoJapan
| | - Shiro Maeda
- Department of Advanced Genomic and Laboratory Medicine, Graduate School of MedicineUniversity of the RyukyusNishihara‐ChoJapan
- Division of Clinical Laboratory and Blood TransfusionUniversity of the Ryukyus HospitalNishihara‐ChoJapan
| |
Collapse
|
50
|
Wen J, Zhao B, Yang Z, Erus G, Skampardoni I, Mamourian E, Cui Y, Hwang G, Bao J, Boquet-Pujadas A, Zhou Z, Veturi Y, Ritchie MD, Shou H, Thompson PM, Shen L, Toga AW, Davatzikos C. The genetic architecture of multimodal human brain age. Nat Commun 2024; 15:2604. [PMID: 38521789 PMCID: PMC10960798 DOI: 10.1038/s41467-024-46796-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 03/06/2024] [Indexed: 03/25/2024] Open
Abstract
The complex biological mechanisms underlying human brain aging remain incompletely understood. This study investigated the genetic architecture of three brain age gaps (BAG) derived from gray matter volume (GM-BAG), white matter microstructure (WM-BAG), and functional connectivity (FC-BAG). We identified sixteen genomic loci that reached genome-wide significance (P-value < 5×10-8). A gene-drug-disease network highlighted genes linked to GM-BAG for treating neurodegenerative and neuropsychiatric disorders and WM-BAG genes for cancer therapy. GM-BAG displayed the most pronounced heritability enrichment in genetic variants within conserved regions. Oligodendrocytes and astrocytes, but not neurons, exhibited notable heritability enrichment in WM and FC-BAG, respectively. Mendelian randomization identified potential causal effects of several chronic diseases on brain aging, such as type 2 diabetes on GM-BAG and AD on WM-BAG. Our results provide insights into the genetics of human brain aging, with clinical implications for potential lifestyle and therapeutic interventions. All results are publicly available at https://labs.loni.usc.edu/medicine .
Collapse
Affiliation(s)
- Junhao Wen
- Laboratory of AI and Biomedical Science (LABS), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA.
| | - Bingxin Zhao
- Department of Statistics and Data Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhijian Yang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guray Erus
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ioanna Skampardoni
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth Mamourian
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yuhan Cui
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Gyujoon Hwang
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jingxuan Bao
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Zhen Zhou
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yogasudha Veturi
- Department of Biobehavioral Health and Statistics, Penn State University, University Park, PA, USA
| | - Marylyn D Ritchie
- Department of Genetics and Institute for Biomedical Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Haochang Shou
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Marina del Rey, CA, USA
| | - Li Shen
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Arthur W Toga
- Laboratory of Neuro Imaging (LONI), Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, USA
| | - Christos Davatzikos
- Artificial Intelligence in Biomedical Imaging Laboratory (AIBIL), Center for AI and Data Science for Integrated Diagnostics (AI2D), Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|