1
|
Zhang L, Feng B, Liu Z, Liu Y. Educational attainment, body mass index, and smoking as mediators in kidney disease risk: a two-step Mendelian randomization study. Ren Fail 2025; 47:2476051. [PMID: 40069100 PMCID: PMC11899219 DOI: 10.1080/0886022x.2025.2476051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/18/2025] [Accepted: 02/23/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Educational attainment (EA) has been linked to various health outcomes, including kidney disease (KD). However, the underlying mechanisms remain unclear. This study aims to assess the causal relationship between EA and KD and quantify the mediation effects of modifiable risk factors using a Mendelian randomization (MR) approach. METHODS We performed a two-sample MR analysis utilizing summary statistics from large-scale European genome-wide association studies (GWAS). EA (NGWAS = 766,345) was used as the exposure, and KD (Ncase/Ncontrol= 5,951/212,871) was the outcome. A two-step MR method was applied to identify and quantify the mediation effects of 24 candidate risk factors. RESULTS Each additional 4.2 years of genetically predicted EA was associated with a 32% reduced risk of KD (odds ratio [OR] 0.68; 95% confidence interval [CI] 0.56, 0.83). Among the 24 candidate risk factors, body mass index (BMI) mediated 21.8% of this protective effect, while smoking heaviness mediated 18.7%. CONCLUSIONS This study provides robust evidence that EA exerts a protective effect against KD, partially mediated by BMI and smoking. These findings highlight the potential for targeted public health interventions aimed at mitigating obesity and smoking-related risks to reduce KD incidence, particularly among individuals with lower educational attainment.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Baiyu Feng
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Zhiwen Liu
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| | - Yu Liu
- Department of Nephrology, The Second Xiangya Hospital at Central South University, Hunan Key Laboratory of Kidney Disease and Blood Purification, Changsha, Hunan, China
| |
Collapse
|
2
|
Dinter KA, Aurich S, Müller L, Ghosh A, Noé F, Wolfrum C, Blüher M, Hoffmann A, Kovacs P, Keller M. Sarcospan, a candidate gene of fat distribution, may affect lipid storage in adipocytes. Mol Cell Endocrinol 2025:112602. [PMID: 40490051 DOI: 10.1016/j.mce.2025.112602] [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: 02/27/2025] [Revised: 05/02/2025] [Accepted: 06/07/2025] [Indexed: 06/11/2025]
Abstract
BACKGROUND AND AIMS Genetic and epigenetic variations in the Sarcospan (SSPN) gene are associated with parameters of fat distribution (body mass index, waist-to-hip ratio), glucose homeostasis and adipocyte size in human potentially by affecting adipogenesis. This study aims at clarifying the impact of SSPN on adipogenesis, particularly focusing on its promoter methylation. MATERIALS AND METHODS Immortalized murine epididymal preadipocytes were transfected with fluorescence-marked plasmids coding for DNMT3a, CRISPR/dCas9-Suntag and vectors carrying guide RNAs complementary to the transcription start site region and differentiated to mature adipocytes. We performed siRNA-mediated Sspn knockdown in epididymal preadipocytes, measured target DNA methylation using pyrosequencing and quantified transcriptional changes of Sspn and adipogenic genes by qPCR. Additionally, we correlated SSPN mRNA values and clinical characteristics from a large human adipose tissue biobank (Leipzig Obesity Biobank). RESULTS Epigenetic editing of the Sspn regulatory region in preadipocytes resulted in a significant increase (up to 35 %) in DNA promoter methylation throughout adipocyte differentiation but showed only minor effects on Sspn expression and fat storage. Though siRNA knockdown could also not contribute to understand the role of Sspn in a 2D adipogenesis model, large-scale correlation analyses still indicate the gene to be a key player in fat distribution and glucose homeostasis. CONCLUSIONS Although the epigenetic downregulation of Sspn showed only marginal effects on adipogenesis, associations of SSPN expression in human adipose tissue with parameters of fat distribution and glucose homeostasis make it a promising candidate for further studies addressing metabolic processes in adipose tissue.
Collapse
Affiliation(s)
- Katharina Anastasia Dinter
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig 04103, Germany
| | - Samantha Aurich
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig 04103, Germany; Department of Animal Physiology, Faculty of Life Sciences, University of Leipzig, Leipzig 04103, Germany
| | - Luise Müller
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig 04103, Germany
| | - Adhideb Ghosh
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach 8092, Switzerland
| | - Falko Noé
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach 8092, Switzerland
| | - Christian Wolfrum
- Institute of Food, Nutrition and Health, ETH Zürich, Schwerzenbach 8092, Switzerland
| | - Matthias Blüher
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig 04103, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig and University Hospital Leipzig, Leipzig 04103, Germany
| | - Anne Hoffmann
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig and University Hospital Leipzig, Leipzig 04103, Germany
| | - Peter Kovacs
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig 04103, Germany; Deutsches Zentrum für Diabetesforschung e.V., Neuherberg 85764, Germany
| | - Maria Keller
- Medical Department III - Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, Leipzig 04103, Germany; Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig and University Hospital Leipzig, Leipzig 04103, Germany.
| |
Collapse
|
3
|
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
|
4
|
Cifarelli V. Lipedema: Progress, Challenges, and the Road Ahead. Obes Rev 2025:e13953. [PMID: 40425048 DOI: 10.1111/obr.13953] [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: 01/31/2025] [Revised: 04/11/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025]
Abstract
INTRODUCTION Lipedema is a chronic and progressive disease that predominantly affects women, characterized by a disproportionate increase in subcutaneous adipose tissue (AT), particularly in the lower limbs. It is associated with significant physical disability, chronic pain, thromboembolism, and psychosocial distress. Despite its profound impact on women's health and quality of life, lipedema remains underrecognized and insufficiently studied, with an estimated prevalence of approximately 10% among women worldwide. Although the exact etiology of lipedema remains unclear, emerging evidence suggests a multifactorial origin involving genetic predisposition, hormonal influences, and vascular dysfunction-all contributing to its development and progression. Current therapeutic options provide only partial symptom relief and remain noncurative, highlighting the urgent need for expanded research and improved management strategies. METHODS A systematic review was conducted to assess the current understanding of lipedema pathophysiology and current treatment options. Research articles were sourced from PubMed, Web of Science, ScienceDirect, and Scopus databases. Over 100 studies were incorporated. RESULTS This review provides a comprehensive overview of lipedema, encompassing its clinical features, pathophysiological mechanisms, diagnostic challenges, and current treatment modalities. Additionally, the review discusses whether the molecular and metabolic differences between abdominal and femoral AT depots mirror those observed in classical obesity. CONCLUSIONS Multidisciplinary, research-informed care is essential for managing lipedema, combining conservative therapies, tailored exercise, and liposuction for advanced cases. More research to better understand the underlying pathophysiology is critical to developing targeted treatments, improving diagnostic accuracy, and informing standardized, evidence-based care.
Collapse
Affiliation(s)
- Vincenza Cifarelli
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
5
|
Kitaba NT, Østergaard TM, Lønnebotn M, Accordini S, Real FG, Malinovschi A, Oudin A, Benediktsdottir B, González FJC, Gómez LP, Holm M, Jõgi NO, Dharmage SC, Skulstad SM, Schlünssen V, Svanes C, Holloway JW. Father's adolescent body silhouette is associated with offspring asthma, lung function and BMI through DNA methylation. Commun Biol 2025; 8:796. [PMID: 40410506 PMCID: PMC12102279 DOI: 10.1038/s42003-025-08121-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 04/23/2025] [Indexed: 05/25/2025] Open
Abstract
Boys' pubertal overweight associates with future offspring's asthma and low lung function. To identify how paternal overweight is associated with offspring's DNA methylation (DNAm), we conducted an epigenome-wide association study of father's body silhouette (FBS) at three timepoints (age 8, voice break and 30) and change in FBS between these times, with offspring DNAm, in the RHINESSA cohort (N = 339). We identified 2005 differentially methylated cytosine-phosphate-guanine (dmCpG) sites (FDR < 0.05), including dmCpGs associated with offspring asthma (119), lung function (178) and BMI (291). Voice break FBS associated with dmCpGs in loci including KCNJ10, FERMT1, NCK2 and WWP1. Change in FBS across sexual maturation associated with DNAm at loci including NOP10, TRRAP, EFHD1, MRPL17 and NORD59A;ATP5B and showed strong correlation in reduced gene expression in loci NAP1L5, ATP5B, ZNF695, ZNF600, VTRNA2-1, SOAT2 and AGPAT2. We identified 24 imprinted genes including: VTRNA2-1, BLCAP, WT1, NAP1L5 and PTPRN2. Identified pathways relate to lipid and glucose metabolism and adipogenesis. Father's overweight at puberty and during reproductive maturation was strongly associated with offspring DNA, suggesting a key role for epigenetic mechanisms in intergenerational transfer from father to offspring in humans. The results support an important vulnerability window in male puberty for future offspring health.
Collapse
Grants
- We thank all the study participants, fieldworkers and scientists in RHINESSA, Co-ordination of the RHINESSA study has received funding from the Research Council of Norway (Grants No. 274767, 214123, 228174, 230827 and 273838), ERC StG project BRuSH #804199, the European Union's Horizon 2020 research and innovation programme under grant agreement No. 633212 (the ALEC Study), the Bergen Medical Research Foundation, and the Western Norwegian Regional Health Authorities (Grants No. 912011, 911892 and 911631). Study centres have further received local funding from the following: Bergen: the above grants for study establishment and co-ordination, and, in addition, World University Network (REF and Sustainability grants), Norwegian Labour Inspection, and the Norwegian Asthma and Allergy Association. Albacete and Huelva: Sociedad Española de Patología Respiratoria (SEPAR) Fondo de Investigación Sanitaria (FIS PS09). Gøteborg, Umeå and Uppsala: the Swedish Heart and Lung Foundation, the Swedish Asthma and Allergy Association. Reykjavik: Iceland University. Melbourne: National Health and Medical Research Council (NHMRC) of Australia (research grants 299901 and 1021275). Tartu: the Estonian Research Council (Grant No. PUT562). Århus: The Danish Wood Foundation (Grant No. 444508795), the Danish Working Environment Authority (Grant No. 20150067134), Aarhus University (PhD scholarship).
Collapse
Affiliation(s)
- Negusse Tadesse Kitaba
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Toril Mørkve Østergaard
- Department of Global Public Health and Primary Care, Centre for International Health, University of Bergen, Bergen, Norway
| | - Marianne Lønnebotn
- Department of Health and Caring Sciences, Western Norway University of Applied Sciences, Bergen, Norway
| | - Simone Accordini
- Unit of Epidemiology and Medical Statistics, Department of Diagnostics and Public Health, University of Verona, Verona, Italy
| | | | - Andrei Malinovschi
- Department of Medical Sciences: Clinical Physiology, Uppsala University, Uppsala, Sweden
| | - Anna Oudin
- Section of Sustainable Health, Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | - Bryndis Benediktsdottir
- Department of Allergy, Respiratory Medicine and Sleep, Landspitali University Hospital, Reykjavik, Iceland Faculty of Medicine, University of Iceland, Landspitali, Iceland
| | | | | | - Mathias Holm
- Occupational and Environmental Medicine, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Nils Oskar Jõgi
- Department of Medical Sciences: Clinical Physiology, Uppsala University, Uppsala, Sweden
| | - Shyamali C Dharmage
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, University of Melbourne, Melbourne, Australia
| | - Svein Magne Skulstad
- Department of Global Public Health and Primary Care, Centre for International Health, University of Bergen, Bergen, Norway
| | - Vivi Schlünssen
- Department of Public Health, Research Unit for Environment, Work and Health, Danish Ramazzini Centre, Aarhus University Denmark, Aarhus, Denmark
| | - Cecilie Svanes
- Department of Global Public Health and Primary Care, Centre for International Health, University of Bergen, Bergen, Norway.
- Department of Occupational Medicine, Haukeland University Hospital, Bergen, Norway.
| | - John W Holloway
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
- NIHR Southampton Biomedical Research Centre, University Hospitals Southampton, Southampton, UK
| |
Collapse
|
6
|
Kim BR, Kim G, Jin SP, Choi CW, Kim J, Park H. Causal association between polyunsaturated fatty acids and acne: a two-sample Mendelian randomization study. Br J Dermatol 2025; 192:1106-1114. [PMID: 39936505 DOI: 10.1093/bjd/ljaf052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Revised: 02/03/2025] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND Observational studies have demonstrated a close association between polyunsaturated fatty acids (PUFAs) and acne. However, the findings of clinical trials have been inconsistent, leaving the causal relationship between PUFAs and acne unclear. OBJECTIVES To investigate the causal association between genetically proxied PUFAs and acne risk. METHODS Mendelian randomization (MR) was performed using single nucleotide polymorphisms associated with PUFAs as instrumental variables. The causal associations between PUFAs and acne were estimated among 115 006 UK Biobank participants and 363 927 participants of Finnish descent. RESULTS Genetically predicted docosahexaenoic acid (DHA) levels [β = -0.303, 95% confidence interval (CI) -0.480 to -0.126; P = 7.74 × 10-4] and its percentage to total fatty acids (β = -0.402, 95% CI -0.651 to -0.258; P = 5.91 × 10-6) showed a significant causal association with a decreased risk of acne. Conversely, genetically predicted percentages of linoleic acid (LA) in total fatty acids (β = 0.768, 95% CI 0.411-0.126; P = 2.87 × 10-4) and omega-6 : omega-3 ratio (β = 0.373, 95% CI 0.142-0.604; P = 4.48 × 10-3) were robustly associated with an increased risk of acne. These effects were attenuated after excluding a genetic variant of rs174528 located upstream of FADS1, highlighting the biologic link between FADS1 and delta-5 desaturase activity. Multivariable MR analysis indicated that PUFAs were causally associated with acne, independent of body mass index. CONCLUSIONS Our study indicates that high DHA levels and their ratios to total fatty acids have causal protective effects against acne, while high LA levels and omega-6 : omega-3 ratio are associated with increased acne risk. This association was largely attributable to the influence of genetic variants related to FADS1.
Collapse
Affiliation(s)
- Bo Ri Kim
- Department of Dermatology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Gahyun Kim
- Precision Medicine Center, Future Innovation Research Division, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Seon-Pil Jin
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Dermatology, Seoul National University Hospital, Seoul, Republic of Korea
| | - Chong Won Choi
- Department of Dermatology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jinho Kim
- Precision Medicine Center, Future Innovation Research Division, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Genomic Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
- Department of Laboratory Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Hyunsun Park
- Department of Dermatology, Seoul National University College of Medicine, Seoul, Republic of Korea
- Department of Dermatology, Seoul National University Hospital, Seoul, Republic of Korea
| |
Collapse
|
7
|
Garza AL, Blangero J, Lee M, Bauer CX, Czerwinski SA, Choh AC. Endophenotype-Informed Association Analyses for Liver Fat Accumulation and Metabolic Dysfunction in the Fels Longitudinal Study. Int J Mol Sci 2025; 26:4812. [PMID: 40429953 PMCID: PMC12112654 DOI: 10.3390/ijms26104812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2025] [Revised: 05/09/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025] Open
Abstract
The identification of causal genomic regions for liver fat accumulation in the context of metabolic dysfunction remains a challenging goal. This study aimed to identify potential endophenotypes for liver fat content and employ them in bivariate linkage searches for pleiotropic genetic regions where targeted association analysis is more likely to reveal significant variants. Multiple metabolic risk and adiposity distribution traits were assessed using the endophenotype ranking value. The top-ranked endophenotypes were then used in a bivariate linkage analysis, paired with liver fat content. Quantitative trait loci (QTLs) identified as significant or suggestive were targeted for measured genotype association analyses. The highest-ranked endophenotypes for liver fat accumulation were insulin resistance (IR), visceral adipose tissue (VAT), and high-density lipoprotein cholesterol (HDL-C). The univariate linkage analysis for liver fat content identified one significant QTL at chromosome 17p13.2 (Logarithm of odds score (LOD) = 2.90, p = 1.29 × 10-4). The bivariate linkage analysis pairing liver fat with IR and VAT improved the localization of two suggestive QTLs at 13q21.31 (LOD = 2.11, p = 9.03 × 10-4), and 6q21 (LOD = 2.35, p = 5.07 × 10-4), respectively. Targeted association analyses within the -1-LOD score regions of these QTLs revealed 17 marginally significant single nucleotide polymorphisms (SNPs) associated with liver fat content or its combination with the selected endophenotypes. The endophenotype-informed linkage analysis successfully identified regions suitable for the targeted association analysis of liver fat content, either alone or in combination with IR or VAT, leading to the discovery of marginally significant variants with potential for future functional studies.
Collapse
Affiliation(s)
- Ariana L. Garza
- School of Public Health, UT Health Science Center, Brownsville, TX 78520, USA
| | - John Blangero
- School of Medicine, South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley, Brownsville, TX 78520, USA
| | - Miryoung Lee
- School of Public Health, Division of Epidemiology, Human Genetics and Environmental Sciences, UT Health Science Center, Brownsville, TX 78520, USA; (M.L.); (A.C.C.)
| | - Cici X. Bauer
- School of Public Health, Division of Biostatistics, UT Health Science Center, Houston, TX 77030, USA
| | - Stefan A. Czerwinski
- School of Health and Rehabilitation Sciences, College of Medicine, Ohio State University, Columbus, OH 43004, USA
| | - Audrey C. Choh
- School of Public Health, Division of Epidemiology, Human Genetics and Environmental Sciences, UT Health Science Center, Brownsville, TX 78520, USA; (M.L.); (A.C.C.)
| |
Collapse
|
8
|
Metz S, Belanich JR, Claussnitzer M, Kilpeläinen TO. Variant-to-function approaches for adipose tissue: Insights into cardiometabolic disorders. CELL GENOMICS 2025; 5:100844. [PMID: 40185091 DOI: 10.1016/j.xgen.2025.100844] [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: 10/31/2024] [Revised: 02/14/2025] [Accepted: 03/12/2025] [Indexed: 04/07/2025]
Abstract
Genome-wide association studies (GWASs) have identified thousands of genetic loci associated with cardiometabolic disorders. However, the functional interpretation of these loci remains a daunting challenge. This is particularly true for adipose tissue, a critical organ in systemic metabolism and the pathogenesis of various cardiometabolic diseases. We discuss how variant-to-function (V2F) approaches are used to elucidate the mechanisms by which GWAS loci increase the risk of cardiometabolic disorders by directly influencing adipose tissue. We outline GWAS traits most likely to harbor adipose-related variants and summarize tools to pinpoint the putative causal variants, genes, and cell types for the associated loci. We explain how large-scale perturbation experiments, coupled with imaging and multi-omics, can be used to screen variants' effects on cellular phenotypes and how these phenotypes can be tied to physiological mechanisms. Lastly, we discuss the challenges and opportunities that lie ahead for V2F research and propose a roadmap for future studies.
Collapse
Affiliation(s)
- Sophia Metz
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Jonathan Robert Belanich
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Melina Claussnitzer
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Programs in Metabolism and Medical & Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Genomic Medicine, Endocrine Division, Massachusetts General Hospital, Harvard Medical School, Cambridge, MA 02142, USA
| | - Tuomas Oskari Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark; The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
9
|
Jiang B, Wei X, Cao X, Zheng C. Insights into modifiable risk factors of retinal vascular occlusion: A Mendelian randomization study. Medicine (Baltimore) 2025; 104:e41752. [PMID: 40324241 PMCID: PMC12055163 DOI: 10.1097/md.0000000000041752] [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: 03/12/2024] [Revised: 01/11/2025] [Accepted: 02/14/2025] [Indexed: 05/07/2025] Open
Abstract
Understanding the etiological risk factors for retinal vascular occlusion (RVO) is critical for prevention and treatment. While the effects of cardiovascular events, hypertension, glaucoma, obesity and glycemic risk factors on RVO are still controversial. This study employed two-sample Mendelian randomization (MR) analysis to investigate these causal risk factors. Single-nucleotide polymorphisms (SNPs) were used as instrumental variables (IVs). Genetic instruments for hypertension, glaucoma, obesity, cardiovascular events and glycemic risk factors were obtained from published genome-wide association studies (GWASs). Summary-level data for RVO and hypertension were obtained from the FinnGen consortium. MR analysis primarily utilized the inverse variance weighted (IVW) method, with MR-Egger and weighted median as supplementary approaches. Multivariable MR (MVMR) adjusting for hypertension or glaucoma of RVO were conducted. Heterogeneity was assessed using Cochrane's Q test and I2, while MR-Egger intercept and MR-PRESSO tested horizontal pleiotropy. All MR analyses were performed within R software (4.1.3) using the R packages "TwoSampleMR" and "MR-PRESSO." Genetic instruments for hypertension and glaucoma were significantly associated with RVO risk. A one-standard deviation (SD) increase in hypertension was associated with a higher risk of RVO [OR = 1.577, 95% CI = (1.342, 1.854), P < .001], while a one-SD increase in the log odds of genetically predicted glaucoma was associated with a higher risk of RVO [OR = 1.24, 95% CI = (1.115, 1.379), P < .001]. Meanwhile, hypertension and glaucoma were still significant in multivariable MR. There was not sufficient evidence to suggest cardiovascular events and obesity were associated with RVO risk. This MR study provided genetic evidence supporting that hypertension and glaucoma were causally associated with the risk of RVO. It may help guide clinical decisions in the management of RVO patients with hypertension and glaucoma.
Collapse
Affiliation(s)
- Bingcai Jiang
- Department of Ophthalmology, Guizhou Provincial People’s Hospital, Guizhou, China
| | - Xin Wei
- Department of Ophthalmology, The People’s Hospital of Tongliang District, Chongqing, China
| | - Xiaochuan Cao
- Department of Ophthalmology, The People’s Hospital of Tongliang District, Chongqing, China
| | - Changwei Zheng
- Department of Ophthalmology, The People’s Hospital of Tongliang District, Chongqing, China
| |
Collapse
|
10
|
Lecoutre S, Rebière C, Maqdasy S, Lambert M, Dussaud S, Abatan JB, Dugail I, Gautier EL, Clément K, Marcelin G. Enhancing adipose tissue plasticity: progenitor cell roles in metabolic health. Nat Rev Endocrinol 2025; 21:272-288. [PMID: 39757324 DOI: 10.1038/s41574-024-01071-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/27/2024] [Indexed: 01/07/2025]
Abstract
Adipose tissue demonstrates considerable plasticity and heterogeneity, enabling metabolic, cellular and structural adaptations to environmental signals. This adaptability is key for maintaining metabolic homeostasis. Impaired adipose tissue plasticity can lead to abnormal adipose tissue responses to metabolic cues, which contributes to the development of cardiometabolic diseases. In chronic obesity, white adipose tissue undergoes pathological remodelling marked by adipocyte hypertrophy, chronic inflammation and fibrosis, which are linked to local and systemic insulin resistance. Research data suggest that the capacity for healthy or unhealthy white adipose tissue remodelling might depend on the intrinsic diversity of adipose progenitor cells (APCs), which sense and respond to metabolic cues. This Review highlights studies on APCs as key determinants of adipose tissue plasticity, discussing differences between subcutaneous and visceral adipose tissue depots during development, growth and obesity. Modulating APC functions could improve strategies for treating adipose tissue dysfunction and metabolic diseases in obesity.
Collapse
Affiliation(s)
- Simon Lecoutre
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France.
| | - Clémentine Rebière
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Salwan Maqdasy
- Department of Medicine, Karolinska Institutet Hospital, Stockholm, Sweden
| | - Mélanie Lambert
- Institut National de la Santé et de la Recherche Médicale, Bobigny, France
- Labex Inflamex, Université Sorbonne Paris Nord, Alliance Sorbonne Paris Cité, Bobigny, France
| | - Sébastien Dussaud
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Jimon Boniface Abatan
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Isabelle Dugail
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Emmanuel L Gautier
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France
| | - Karine Clément
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France.
- Department of Nutrition, Pitie-Salpêtriere Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.
| | - Geneviève Marcelin
- Nutrition and Obesities: Systemic Approach Research Group, Nutriomics, Sorbonne Université, INSERM, Paris, France.
| |
Collapse
|
11
|
Ene P, Svensson MK, Strand R, Kullberg J, Ahlström H, Larsson A, Lind L. Causal effects of obesity on estimated glomerular filtration rate: a Mendelian randomization and image data analysis study. Clin Kidney J 2025; 18:sfaf116. [PMID: 40357501 PMCID: PMC12067075 DOI: 10.1093/ckj/sfaf116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Indexed: 05/15/2025] Open
Abstract
Background Obesity has been associated with onset and progression of chronic kidney disease (CKD) but causal relationship remains uncertain. This study investigated how obesity causally affects estimated glomerular filtration rate. Methods Cross-sectional and magnetic resonance imaging (MRI) data analyses were performed within the Prospective Investigation of Obesity, Energy, and Metabolism (POEM) study (502 participants, all aged 50 years). Additionally Mendelian randomization was performed using published summary data. Outcomes were creatinine- and cystatin C-based eGFR. Body mass index (BMI) and waist circumference (WC) were used as exposure variables in the cross-sectional and Mendelian randomization analyses. In the imaging data analyses, eGFR was regressed non-parametrically on tissue volume for each 3D voxel and visualized as a correlation "Imiomics" map. Results Negative correlations were shown between cystatin C-based eGFR and BMI [beta = -0.190 (95% CI: -0.280 to -0.100)] and WC [beta = -0.160 (95% CI: -0.250 to -0.060)] in an adjusted model. In contrast, a positive association was found for creatinine-based eGFR [BMI beta = 1.20 (95% CI: 0.030 to 0.210) and WC beta = 0.160 (95% CI: 0.070 to 0.260)]. Similar patterns were found using MRI analysis (Imiomics map). Mendelian randomization implied a negative causal effect of obesity-related measures on cystatin C-based eGFR [BMI beta = -0.031 (95% CI: -0.037 to -0.026) and WC beta = -0.038 (95% CI: -0.045 to -0.031)], but no statistically significant effect was found for creatinine-based eGFR. Conclusion This study suggests a causal negative effect of obesity on cystatin C-based, but not creatinine-based eGFR. These findings warrant further research regarding estimations of kidney function when assessing obesity and CKD.
Collapse
Affiliation(s)
- Patrik Ene
- Department of Medical Sciences Renal Medicine, Uppsala University Hospital, Uppsala University
| | - Maria K Svensson
- Department of Medical Sciences Renal Medicine, Uppsala University Hospital, Uppsala University
- Uppsala Clinical Research Centre, Uppsala
| | - Robin Strand
- Department of Information Technology, Uppsala University
| | - Joel Kullberg
- Department of Radiology, Uppsala University Hospital, Uppsala University
| | - Håkan Ahlström
- Department of Radiology, Uppsala University Hospital, Uppsala University
| | - Anders Larsson
- Department of Clinical Chemistry, Uppsala University Hospital, Uppsala University
| | - Lars Lind
- Department of Medical Sciences, Uppsala University Hospital, Uppsala University, all from Sweden
| |
Collapse
|
12
|
Zhao SS, Mackie SL, Larsson SC, Burgess S, Yuan S. Modifiable risk factors and inflammation-related proteins in polymyalgia rheumatica: genome-wide meta-analysis and Mendelian randomization. Rheumatology (Oxford) 2025; 64:3012-3018. [PMID: 38788669 PMCID: PMC7616751 DOI: 10.1093/rheumatology/keae308] [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: 04/21/2024] [Revised: 05/13/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
OBJECTIVE PMR is an age-related inflammatory disease of unknown cause. We aimed to identify potentially modifiable risk factors and therapeutic targets for preventing or treating PMR. METHODS We meta-analysed genetic association data from 8156 cases of PMR (defined using diagnostic codes and self-report) and 416 495 controls of European ancestry from the UK Biobank and FinnGen. We then performed Mendelian randomization analyses to estimate the association between eight modifiable risk factors (using data from up to 1.2 million individuals) and 65 inflammation-related circulating proteins (up to 55 792 individuals), using the inverse variance weighted and pleiotropy robust methods. RESULTS We identified three novel genome-wide significant loci in the IL1R1, NEK6 and CCDC88B genes and confirmation of previously described associations with HLA-DRB1 and ANKRD55. Genetically predicted smoking intensity (OR 1.32; 95%CI 1.08-1.60; P = 0.006) and visceral adiposity (OR 1.22; 95%CI 1.10-1.37; P = 3.10 × 10-4) were associated with PMR susceptibility. Multiple circulating proteins related to IL-1 family signalling were associated with PMR. IL-1 receptor-like 2, also known as IL-36 receptor (OR 1.25; P = 1.89 × 10-32), serum amyloid A2 (OR 1.06, 9.91 × 10-10) and CXCL6 (OR 1.09, P = 4.85 × 10-7) retained significance after correction for multiple testing. CONCLUSION Reducing smoking and visceral adiposity at a population level might reduce incidence of PMR. We identified proteins that may play causal roles in PMR, potentially suggesting new therapeutic opportunities. Further research is needed before these findings are applied to clinical practice.
Collapse
Affiliation(s)
- Sizheng Steven Zhao
- Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Science, School of Biological Sciences, Faculty of Biological Medicine and Health, The University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Sarah L Mackie
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, UK
- National Institute for Health Research Leeds Biomedical Research Centre, Leeds Teaching Hospitals, University of Leeds, Leeds, UK
| | - Susanna C Larsson
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Stephen Burgess
- British Heart Foundation Cardiovascular Epidemiology Unit, Department of Public Health and Primary Care, University of Cambridge, Cambridge, UK
- Heart and Lung Research Institute, University of Cambridge, Cambridge, UK
- Medical Research Council Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Shuai Yuan
- Unit of Cardiovascular and Nutritional Epidemiology, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
13
|
Zhang M, Ward J, Strawbridge RJ, Anderson JJ, Celis-Morales C, Pell JP, Ho FK, Lyall DM. Genetic predisposition to adiposity, and type 2 diabetes: the role of lifestyle and phenotypic adiposity. Eur J Endocrinol 2025; 192:549-557. [PMID: 40315335 PMCID: PMC12056655 DOI: 10.1093/ejendo/lvaf084] [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: 01/21/2025] [Revised: 04/14/2025] [Accepted: 04/23/2025] [Indexed: 05/04/2025]
Abstract
AIMS Genetic predisposition to adiposity is associated with type 2 diabetes (T2D), even in the absence of phenotypic adiposity (obesity and central obesity). We aimed to quantify the overall contribution of obesity and modifiable lifestyle factors to the association between genetic predisposition to adiposity and the development of T2D. METHODS This prospective cohort study involved 220 703 White British participants from the UK Biobank. It examined the associations between genetic predisposition to adiposity [body mass index polygenic risk (BMI-PRS) and waist-hip ratio polygenic risk (WHR-PRS)] and incident T2D, as well as interactions and mediation via lifestyle factors (diet quality, physical activity levels, total energy intake, sleep duration, and smoking and alcohol intake) and phenotypic adiposity. RESULTS People with high phenotypic adiposity and high adiposity PRS values (>1 SD above the mean) had the highest risk of incident T2D (versus non-obese/central obese and non-high PRS). This was the case for BMI-PRS [hazard ratio (HR) = 3.72] and WHR-PRS (HR = 4.17). Lifestyle factors explained 30.5% of the BMI-PRS/T2D association (2.0% mediation; 28.5% effect modification), and lifestyle and obesity together explained 92.1% (78.8% mediation; 13.3% effect modification). Lifestyle factors explained 20.4% of the WHR-PRS/T2D association (3.4% mediation; 17.0% effect modification), and lifestyle and central obesity together explained 72.8% (41.1% mediation; 31.7% effect modification). CONCLUSIONS Whilst phenotypic adiposity explains a large proportion of the association between BMI-PRS/WHR-PRS and T2D, modifiable lifestyle factors also make contributions. Promoting healthy lifestyles among people prone to adiposity is important in reducing the global burden of T2D.
Collapse
Affiliation(s)
- Mengrong Zhang
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8TB, United Kingdom
| | - Joey Ward
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8TB, United Kingdom
| | - Rona J Strawbridge
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8TB, United Kingdom
- Department of Medicine Solna, Karolinska Institute, Stockholm 17177, Sweden
| | - Jana J Anderson
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8TB, United Kingdom
| | - Carlos Celis-Morales
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8TB, United Kingdom
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow G12 8TA, United Kingdom
- Human Performance Lab, Education, Physical Activity, and Health Research Unit, Universidad Católica del Maule, Talca 115 3745, Chile
- Centro de Investigación en Medicina de Altura (CEIMA), Universidad Arturo Prat, Iquique 1100012, Chile
| | - Jill P Pell
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8TB, United Kingdom
| | - Frederick K Ho
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8TB, United Kingdom
| | - Donald M Lyall
- School of Health and Wellbeing, University of Glasgow, Glasgow G12 8TB, United Kingdom
| |
Collapse
|
14
|
Han K, Wang X, Chen S, Niu X, Wang Y, Xiang J, Ru N, Liu M, Chai N, Linghu E. Association of Genetically Predicted Obesity and Stool Frequency: Evidence From an Observational and Mendelian Randomization Study. J Neurogastroenterol Motil 2025; 31:267-275. [PMID: 40205902 PMCID: PMC11986650 DOI: 10.5056/jnm23178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 06/16/2024] [Accepted: 08/13/2024] [Indexed: 04/11/2025] Open
Abstract
Background/Aims Obesity is associated with several gastrointestinal (GI) disorders and has been identified as a potential risk factor for various GI symptoms. Bowel frequency is an important indicator of bowel function. However, the causal link between obesity and gastrointestinal motility remains uncertain. This study aims to determine the causal effect of overall and central obesity on stool frequency. Methods Four obesity-related anthropometric indicators-body mass index, body fat percentage, waist circumference (WC), and waist-to-hip ratio (WHR)-were investigated. Individual-level baseline information from the UK Biobank was used to explore observational associations between obesity and stool frequency. Additionally, summary-level data from published genome-wide association studies were subjected to two-sample Mendelian randomization (MR) analyses to examine causal associations. Results For all 4 indicators of obesity, higher levels of obesity were associated with more frequent bowel movements after adjusting for demographic characteristics, lifestyle, and dietary factors. After rigorous screening, 482 body mass index single nucleotide polymorphisms (SNPs), 7 body fat percentage SNPs, 48 WC SNPs, and 287 WHR SNPs were identified as instrument variables for MR analysis. The MR results were generally consistent with observational findings, proving that the associations observed in the overall obesity indicators were causal. For central obesity, the association between WHR and stool frequency remained consistent in both analysis phases, whereas WC showed a multidirectional association. Conclusions Obesity-related anthropometric indicators were causally associated with increased stool frequency in the overall and central obesity groups. Weight loss could be a potential approach to improve gastrointestinal regularity in individuals with obesity.
Collapse
Affiliation(s)
- Ke Han
- Department of Endoscopy, General Hospital of Northern Theater Command, Shenyang, China
- Department of Gastroenterology and Hepatology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiangyao Wang
- Department of Gastroenterology and Hepatology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Shimin Chen
- Institute of Geriatrics, Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatrics Diseases, Second Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiaotong Niu
- Department of Gastroenterology and Hepatology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yan Wang
- Department of Gastroenterology and Hepatology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jingyuan Xiang
- Department of Gastroenterology and Hepatology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Nan Ru
- Department of Gastroenterology and Hepatology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Miao Liu
- Department of Statistics and Epidemiology, Graduate School of Chinese PLA General Hospital, Beijing, China
| | - Ningli Chai
- Department of Gastroenterology and Hepatology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Enqiang Linghu
- Department of Gastroenterology and Hepatology, First Medical Center of Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
15
|
Ramessur R, Saklatvala J, Løset M, Thomas LF, Budu-Aggrey A, Mahil SK, Barker JN, Dand N, Simpson MA, Smith CH. Investigating the Genetic Basis of the Influence of Adiposity on Psoriasis: A Cross-Sectional Study in a Large United Kingdom Population-Based Biobank. J Invest Dermatol 2025:S0022-202X(25)00391-4. [PMID: 40423604 DOI: 10.1016/j.jid.2025.03.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/04/2025] [Accepted: 03/20/2025] [Indexed: 05/28/2025]
Affiliation(s)
- Ravi Ramessur
- St John's Institute of Dermatology, Faculty of Life Sciences & Medicine, School of Basic & Medical Biosciences, King's College London, London, United Kingdom
| | - Jake Saklatvala
- Department of Medical and Molecular Genetics, School of Basic & Medical Biosciences, King's College London, London, United Kingdom
| | - Mari Løset
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway; Department of Orthopaedics, Rheumatology and Dermatology, St. Olavs hospital, Trondheim University Hospital, Trondheim, Norway
| | - Laurent F Thomas
- HUNT Center for Molecular and Clinical Epidemiology, Department of Public Health and Nursing, Norwegian University of Science and Technology, Trondheim, Norway; Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway; BioCore - Bioinformatics Core Facility, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ashley Budu-Aggrey
- MRC Integrative Epidemiology Unit at University of Bristol, Bristol, United Kingdom; Population Health Sciences, Bristol Medical School, Bristol, United Kingdom
| | - Satveer K Mahil
- St John's Institute of Dermatology, Faculty of Life Sciences & Medicine, School of Basic & Medical Biosciences, King's College London, London, United Kingdom
| | - Jonathan N Barker
- St John's Institute of Dermatology, Faculty of Life Sciences & Medicine, School of Basic & Medical Biosciences, King's College London, London, United Kingdom
| | - Nick Dand
- Department of Medical and Molecular Genetics, School of Basic & Medical Biosciences, King's College London, London, United Kingdom
| | - Michael A Simpson
- Department of Medical and Molecular Genetics, School of Basic & Medical Biosciences, King's College London, London, United Kingdom
| | - Catherine H Smith
- St John's Institute of Dermatology, Faculty of Life Sciences & Medicine, School of Basic & Medical Biosciences, King's College London, London, United Kingdom.
| |
Collapse
|
16
|
Seo J, Kim G, Park S, Lee A, Liang L, Park T, Chung W. Assessing the causal effects of type 2 diabetes and obesity-related traits on COVID-19 severity. Hum Genomics 2025; 19:43. [PMID: 40264243 PMCID: PMC12016339 DOI: 10.1186/s40246-025-00747-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND Type 2 diabetes (T2D) and obesity-related traits are highly comorbid with coronavirus disease 2019 (COVID-19), but their causal relationships with disease severity remain unclear. While recent Mendelian randomization (MR) studies suggest a causal link between obesity-related traits and COVID-19 severity, findings regarding T2D are inconsistent, particularly when adjusting for body mass index (BMI). This study aims to clarify these relationships. METHODS We applied various MR methods to assess the causal effects of BMI-adjusted T2D (T2DadjBMI) and obesity-related traits (BMI, waist circumference, and waist-hip ratio) on COVID-19 severity. Genetic instruments were obtained from large-scale genome-wide association studies (GWAS), including 898K participants for T2D and 2M for COVID-19 severity. To address potential bias from sample overlap, we conducted large-scale simulations comparing MR results from overlapping and independent samples. RESULTS Our MR analysis identified a significant causal relationship between T2DadjBMI and increased COVID-19 severity (OR = 1.057, 95% CI = 1.012-1.105). Obesity-related traits were also causally associated with COVID-19 severity. Simulations confirmed that MR results remained robust to sample overlap, demonstrating consistency between overlapping and independent datasets. CONCLUSIONS These findings highlight the causal role of T2D and obesity-related traits in COVID-19 severity, emphasizing the need for targeted prevention and management strategies for high-risk populations. The robustness of our MR analysis, even in the presence of sample overlap, strengthens the reliability of these causal inferences.
Collapse
Affiliation(s)
- Jieun Seo
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, 06978, Korea
| | - Gaeun Kim
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, 06978, Korea
| | - Seunghwan Park
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, 06978, Korea
| | - Aeyeon Lee
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, 06978, Korea
| | - Liming Liang
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Taesung Park
- Department of Statistics, Seoul National University, Seoul, 08826, Korea.
| | - Wonil Chung
- Department of Statistics and Actuarial Science, Soongsil University, Seoul, 06978, Korea.
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, 02115, USA.
| |
Collapse
|
17
|
Sun J, Chen L, Zhao P, Bai X, Nie S, Li Y. New insights into causal relationship between serum lipids, obesity, and asthma: a Mendelian randomization study. J Asthma 2025:1-12. [PMID: 40227003 DOI: 10.1080/02770903.2025.2493177] [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: 02/01/2025] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
OBJECTIVE To identify causal risk factors for asthma using a Mendelian randomization (MR) approach. METHODS Genetic variants associated with the exposures at the genome-wide significance level (p < 5 × 10 - 8) were obtained from corresponding genome-wide association studies. Summary-level statistical data for asthma were obtained from the UK Biobank (UKB) and the FinnGen Consortia. Univariate and multivariate MR analyses were performed to clarify causal relationships among obesity, serum lipids, and asthma. Meta-analyses were performed to combine UKB and FinnGen results using a fixed-effects model. RESULTS In FinnGen, the odds for asthma increased for every 1-SD increase in body mass index (BMI; odds ratio [OR] 1.292, p = 1.34 × 10-7), together with body fat percentage (BF%; OR 1.449, p = 4.90×10-3), and total cholesterol level (OR = 0.949, p = 0.027). However, higher BMI and BF% were found to increase the risk for asthma in the multivariate MR analysis. In the UKB, the BMI results were replicated. Meta-analysis revealed that high-density lipoprotein cholesterol could also increase the risk for asthma, although there were no associations with other risk factors included in this study. CONCLUSION This MR study found that genetically predicted higher BF% and BMI could increase the risk for asthma and corroborated some risk factors for asthma from previous MR studies. Moreover, the results suggest that higher BMI and BF% could serve as independent risk factors for asthma.
Collapse
Affiliation(s)
- Jialiang Sun
- Department of Pediatric Pulmonary Medicine, Children's Medical Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Lanlan Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Peiliang Zhao
- Department of Pediatric Pulmonary Medicine, Children's Medical Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xinquan Bai
- Department of Pediatric Pulmonary Medicine, Children's Medical Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Shu Nie
- Department of Pediatric Cardiology, Children's Medical Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yanan Li
- Department of Pediatric Pulmonary Medicine, Children's Medical Center, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
18
|
Lin E, Yan YT, Chen MH, Yang AC, Kuo PH, Tsai SJ. Gene clusters linked to insulin resistance identified in a genome-wide study of the Taiwan Biobank population. Nat Commun 2025; 16:3525. [PMID: 40229288 PMCID: PMC11997021 DOI: 10.1038/s41467-025-58506-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: 09/27/2024] [Accepted: 03/25/2025] [Indexed: 04/16/2025] Open
Abstract
This pioneering genome-wide association study examined surrogate markers for insulin resistance (IR) in 147,880 Taiwanese individuals using data from the Taiwan Biobank. The study focused on two IR surrogate markers: the triglyceride to high-density lipoprotein cholesterol (TG:HDL-C) ratio and the TyG index (the product of fasting plasma glucose and triglycerides). We identified genome-wide significance loci within four gene clusters: GCKR, MLXIPL, APOA5, and APOC1, uncovering 197 genes associated with IR. Transcriptome-wide association analysis revealed significant associations between these clusters and TyG, primarily in adipose tissue. Gene ontology analysis highlighted pathways related to Alzheimer's disease, glucose homeostasis, insulin resistance, and lipoprotein dynamics. The study identified sex-specific genes associated with TyG. Polygenic risk score analysis linked both IR markers to gout and hyperlipidemia. Our findings elucidate the complex relationships between IR surrogate markers, genetic predisposition, and disease phenotypes in the Taiwanese population, contributing valuable insights to the field of metabolic research.
Collapse
Affiliation(s)
- Eugene Lin
- Department of Genome Sciences, University of Washington, Seattle, WA, USA
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan, ROC
| | - Yu-Ting Yan
- Department of Public Health & Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Mu-Hong Chen
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Department of Psychiatry, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
| | - Albert C Yang
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
| | - Po-Hsiu Kuo
- Department of Public Health & Institute of Epidemiology and Preventive Medicine, National Taiwan University, Taipei, Taiwan, ROC.
- Department of Psychiatry, National Taiwan University Hospital, Taipei, Taiwan, ROC.
| | - Shih-Jen Tsai
- Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan, ROC.
- Department of Psychiatry, College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC.
- Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan, ROC.
| |
Collapse
|
19
|
McCaw ZR, Dey R, Somineni H, Amar D, Mukherjee S, Sandor K, Karaletsos T, Koller D, Aschard H, Smith GD, MacArthur D, O'Dushlaine C, Soare TW. Pitfalls in performing genome-wide association studies on ratio traits. HGG ADVANCES 2025; 6:100406. [PMID: 39818621 PMCID: PMC11808723 DOI: 10.1016/j.xhgg.2025.100406] [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: 10/07/2024] [Revised: 01/08/2025] [Accepted: 01/08/2025] [Indexed: 01/18/2025] Open
Abstract
Genome-wide association studies (GWASs) are often performed on ratios composed of a numerator trait divided by a denominator trait. Examples include body mass index (BMI) and the waist-to-hip ratio, among many others. Explicitly or implicitly, the goal of forming the ratio is typically to adjust for an association between the numerator and denominator. While forming ratios may be clinically expedient, there are several important issues with performing GWAS on ratios. Forming a ratio does not "adjust" for the denominator in the sense of conditioning on it, and it is unclear whether associations with ratios are attributable to the numerator, the denominator, or both. Here we demonstrate that associations arising in ratio GWAS can be entirely denominator driven, implying that at least some associations uncovered by ratio GWAS may be due solely to a putative adjustment variable. In a survey of 10 common ratio traits, we find that the ratio model disagrees with the adjusted model (performing GWAS on the numerator while conditioning on the denominator) at around 1/3 of loci. Using BMI as an example, we show that variants detected by only the ratio model are more strongly associated with the denominator (height), while variants detected by only the adjusted model are more strongly associated with the numerator (weight). Although the adjusted model provides effect sizes with a clearer interpretation, it is susceptible to collider bias. We propose and validate a simple method of correcting for the genetic component of collider bias via leave-one-chromosome-out polygenic scoring.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hugues Aschard
- Institut Pasteur, Université Paris Cité, Department of Computational Biology, Paris, France
| | | | - Daniel MacArthur
- Centre for Population Genomics, Garvan Institute of Medical Research, Sydney, NSW, Australia; Centre for Population Genomics, Murdoch Children's Research Institute, Melbourne, VIC, Australia
| | | | | |
Collapse
|
20
|
Chalitsios CV, Markozannes G, Papagiannopoulos C, Aglago EK, Berndt SI, Buchanan DD, Campbell PT, Cao Y, Chan AT, Dimou N, Drew DA, French AJ, Georgeson P, Giannakis M, Gruber SB, Gunter MJ, Harrison TA, Hoffmeister M, Hsu L, Huang WY, Hullar MAJ, Huyghe JR, Lynch BM, Moreno V, Newton CC, Nowak JA, Obón-Santacana M, Ogino S, Qu C, Schmit SL, Steinfelder RS, Sun W, Thomas CE, Toland AE, Trinh QM, Ugai T, Um CY, Van Guelpen B, Zaidi SH, Murphy N, Peters U, Phipps AI, Tsilidis KK. Waist Circumference, a Body Shape Index, and Molecular Subtypes of Colorectal Cancer: A Pooled Analysis of Four Cohort Studies. Cancer Epidemiol Biomarkers Prev 2025; 34:568-577. [PMID: 39898780 DOI: 10.1158/1055-9965.epi-24-1534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/27/2024] [Accepted: 01/29/2025] [Indexed: 02/04/2025] Open
Abstract
BACKGROUND Waist circumference (WC) and its allometric counterpart, "a body shape index" (ABSI), are risk factors for colorectal cancer; however, it is uncertain whether associations with these body measurements are limited to specific molecular subtypes of the disease. METHODS Data from 2,772 colorectal cancer cases and 3,521 controls were pooled from four cohort studies within the Genetics and Epidemiology of Colorectal Cancer Consortium. Four molecular markers (BRAF mutation, KRAS mutation, CpG island methylator phenotype, and microsatellite instability) were analyzed individually and in combination (Jass types). Multivariable logistic and multinomial logistic models were used to assess the associations of WC and ABSI with overall colorectal cancer risk and, in case-only analyses, to evaluate heterogeneity by molecular subtype, respectively. RESULTS Higher WC (ORper 5 cm = 1.06, 95% confidence interval, 1.04-1.09) and ABSI (ORper 1-SD = 1.07, 95% confidence interval, 1.00-1.14) were associated with elevated colorectal cancer risk. There was no evidence of heterogeneity between the molecular subtypes. No difference was observed regarding the influence of WC and ABSI on the four major molecular markers in proximal colon, distal colon, and rectal cancers, as well as in early- and late-onset colorectal cancers. Associations did not differ in the Jass-type analysis. CONCLUSIONS Higher WC and ABSI were associated with elevated colorectal cancer risk; however, they do not differentially influence all four major molecular mutations involved in colorectal carcinogenesis but underscore the importance of maintaining a healthy body weight in colorectal cancer prevention. IMPACT The proposed results have potential utility in colorectal cancer prevention.
Collapse
Affiliation(s)
| | - Georgios Markozannes
- Department of Hygiene and Epidemiology, University of Ioannina, Ioannina, Greece
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | | | - Elom K Aglago
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | - Sonja I Berndt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Daniel D Buchanan
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Melbourne Medical School, The University of Melbourne, Parkville, Australia
- University of Melbourne Centre for Cancer Research, The University of Melbourne, Parkville, Australia
- Genomic Medicine and Family Cancer Clinic, The Royal Melbourne Hospital, Parkville, Australia
| | - Peter T Campbell
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, New York
| | - Yin Cao
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, St Louis, Missouri
- Alvin J. Siteman Cancer Center at Barnes-Jewish Hospital and Washington University School of Medicine, St. Louis, Missouri
- Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Andrew T Chan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Channing Division of Network Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Niki Dimou
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
| | - David A Drew
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Amy J French
- Division of Laboratory Genetics, Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Peter Georgeson
- Colorectal Oncogenomics Group, Department of Clinical Pathology, Melbourne Medical School, The University of Melbourne, Parkville, Australia
- University of Melbourne Centre for Cancer Research, The University of Melbourne, Parkville, Australia
| | - Marios Giannakis
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Stephen B Gruber
- Department of Medical Oncology and Therapeutics Research and Center for Precision Medicine, City of Hope National Medical Center, Duarte, California
| | - Marc J Gunter
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
| | - Tabitha A Harrison
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Epidemiology, University of Washington School of Public Health, Seattle, Washington
| | - Michael Hoffmeister
- Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Li Hsu
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Biostatistics, University of Washington, Seattle, Washington
| | - Wen-Yi Huang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland
| | - Meredith A J Hullar
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Jeroen R Huyghe
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Brigid M Lynch
- Centre for Epidemiology and Biostatistics, Melbourne School of Population and Global Health, The University of Melbourne, Parkville, Australia
- Cancer Epidemiology Division, Cancer Council Victoria, Melbourne, Australia
| | - Victor Moreno
- Unit of Biomarkers and Suceptibility (UBS), Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Department of Clinical Sciences, Faculty of Medicine and Health Sciences and Universitat de Barcelona Institute of Complex Systems (UBICS), University of Barcelona (UB), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Christina C Newton
- Department of Population Science, American Cancer Society, Atlanta, Georgia
| | - Jonathan A Nowak
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| | - Mireia Obón-Santacana
- Unit of Biomarkers and Suceptibility (UBS), Oncology Data Analytics Program (ODAP), Catalan Institute of Oncology (ICO), L'Hospitalet del Llobregat, Barcelona, Spain
- ONCOBELL Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- Consortium for Biomedical Research in Epidemiology and Public Health (CIBERESP), Madrid, Spain
- Department of Clinical Sciences, Faculty of Medicine and Health Sciences and Universitat de Barcelona Institute of Complex Systems (UBICS), University of Barcelona (UB), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Shuji Ogino
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Program in MPE Molecular Pathological Epidemiology, Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
- Tokyo Medical and Dental University (Institute of Science Tokyo), Tokyo, Japan
| | - Conghui Qu
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Stephanie L Schmit
- Genomic Medicine Institute, Cleveland Clinic, Cleveland, Ohio
- Population and Cancer Prevention Program, Case Comprehensive Cancer Center, Cleveland, Ohio
| | - Robert S Steinfelder
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Wei Sun
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Claire E Thomas
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Amanda E Toland
- Department of Cancer Biology and Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
- Department of Internal Medicine, Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio
| | - Quang M Trinh
- Ontario Institute for Cancer Research, Toronto, Canada
| | - Tomotaka Ugai
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, Boston, Massachusetts
| | - Caroline Y Um
- Department of Population Science, American Cancer Society, Atlanta, Georgia
| | - Bethany Van Guelpen
- Department of Diagnostics and Intervention, Oncology Unit, Umeå University, Umeå, Sweden
- Wallenberg Centre for Molecular Medicine, Umeå University, Umeå, Sweden
| | - Syed H Zaidi
- Ontario Institute for Cancer Research, Toronto, Canada
| | - Neil Murphy
- Nutrition and Metabolism Branch, International Agency for Research on Cancer, Lyon, France
| | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Epidemiology, University of Washington School of Public Health, Seattle, Washington
| | - Amanda I Phipps
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington
- Department of Epidemiology, University of Washington School of Public Health, Seattle, Washington
| | - Konstantinos K Tsilidis
- Department of Hygiene and Epidemiology, University of Ioannina, Ioannina, Greece
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| |
Collapse
|
21
|
Wang Z, Lu Q, Hou S, Zhu H. Genetic causal effects of multi-site chronic pain on post-traumatic stress disorder: Evidence from a two-sample, two-step Mendelian randomization study. Prog Neuropsychopharmacol Biol Psychiatry 2025; 138:111307. [PMID: 40044071 DOI: 10.1016/j.pnpbp.2025.111307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/13/2025] [Accepted: 03/01/2025] [Indexed: 03/09/2025]
Abstract
BACKGROUND Existing evidence supports a correlation between multi-site chronic pain and post-traumatic stress disorder (PTSD), but it is yet to be determined if this correlation is causal and in what direction the causation works. METHODS Applying two-sample Mendelian randomization (MR) analysis to data from available genome-wide association studies in populations of European ancestry, we estimated the causal association between multi-site chronic pain and no pain versus PTSD. Moreover, we used multivariable and mediation MR analysis to assess the mediating effects of 13 lifestyle factors or diseases on the causal relationship between multi-site chronic pain and PTSD. The MR analyses were mainly conducted with the inverse variance weighted (IVW) method, followed by various sensitivity and validation analyses. RESULTS Multi-site chronic pain dramatically increases the risk of developing PTSD (odds ratio [OR]IVW = 2.39, 95 % confidence interval [CI] = 1.72-3.31, p = 2.10 × 10-7), and no pain significantly reduces the risk of developing PTSD (ORIVW = 0.12, 95 % CI = 0.05-0.30, p = 3.14 × 10-6). Multivariable MR found that 13 potential confounding factors do not influence the causal effect of multi-site chronic pain on PTSD. Moreover, body mass index (BMI) (6.98 %), educational attainment (8.79 %), major depressive disorder (MDD) (36.98 %) and insomnia (27.25 %) mediate the causal connection between multi-site chronic pain and PTSD. CONCLUSION Overall, individuals with multi-site chronic pain may be at a higher risk of developing PTSD, and this risk is partially influenced by the pathways involving BMI, educational attainment, MDD, and insomnia. These factors offer potential targets for therapeutic interventions.
Collapse
Affiliation(s)
- Zuxing Wang
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610031, China
| | - Qiao Lu
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610031, China
| | - Shuyu Hou
- Mental Health Center, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Hongru Zhu
- Mental Health Center, West China Hospital of Sichuan University, Chengdu 610041, China; Huaxi Brain Research Center, West China Hospital of Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
22
|
Cheng C, Li Z, Su Y, Sun J, Xu C, Kong X, Sun W. Obesity, Visceral Adipose Tissue, and Essential Hypertension: Evidence From a Mendelian Randomization Study and Mediation Analysis. J Clin Hypertens (Greenwich) 2025; 27:e70045. [PMID: 40259745 PMCID: PMC12012245 DOI: 10.1111/jch.70045] [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: 01/22/2025] [Revised: 03/19/2025] [Accepted: 03/29/2025] [Indexed: 04/23/2025]
Abstract
This study aims to investigate the causal relationship between obesity and essential hypertension, and evaluate the mediation effect of visceral adipose tissue (VAT) by Mendelian randomization (MR) analysis. We included body mass index (BMI), waist circumference (WC), waist-hip ratio (WHR), WC adjusted for BMI (WCadjbmi), and WHR adjusted for BMI (WHRadjbmi) as obesity-related anthropometric traits. In the bidirectional MR analyses, we found that higher BMI (OR, 1.638; p = 3.97 × 10-19), WC (OR, 1.702; p = 1.45 × 10-12), and WHR (OR, 1.863; p = 1.84 × 10-8) were significantly associated with increased risk of essential hypertension, while no evidence of reverse causality was observed. Then, in the two-step MR analyses, all five anthropometric traits had a positive and significant association with VAT mass, especially WC (OR, 2.315; p = 1.00 × 10-210). Meanwhile, higher predicted VAT mass was significantly associated with increased risk of essential hypertension (OR, 1.713; p = 1.18 × 10-38). Furthermore, the mediation analyses revealed that VAT had a significant mediation effect on the causal relationship between obesity-related anthropometric traits and essential hypertension, and mediated proportions in BMI, WC, and WHR were 77.8%, 80.1%, and 41.4%, respectively. Finally, the sensitivity analyses using two other datasets showed a similar result. In conclusion, our results showed that BMI, WC, and WHR have a positive and significant association with increased risk of essential hypertension. Moreover, VAT has a significant mediation effect on the causal relationship between obesity-related anthropometric traits and essential hypertension. Our study provided important statistical evidence suggesting that VAT may play a crucial meditation role in the occurrence and development of obesity-related hypertension.
Collapse
Affiliation(s)
- Chen Cheng
- Department of CardiologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
- Cardiovascular Research CenterThe Affiliated Suzhou Hospital of Nanjing Medical UniversitySuzhou Municipal HospitalGusu SchoolNanjing Medical UniversitySuzhouJiangsuChina
| | - Zheng Li
- Department of CardiologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
- Cardiovascular Research CenterThe Affiliated Suzhou Hospital of Nanjing Medical UniversitySuzhou Municipal HospitalGusu SchoolNanjing Medical UniversitySuzhouJiangsuChina
| | - Yue Su
- Department of CardiologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
- Cardiovascular Research CenterThe Affiliated Suzhou Hospital of Nanjing Medical UniversitySuzhou Municipal HospitalGusu SchoolNanjing Medical UniversitySuzhouJiangsuChina
| | - Jin‐Yu Sun
- Department of CardiologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
- Cardiovascular Research CenterThe Affiliated Suzhou Hospital of Nanjing Medical UniversitySuzhou Municipal HospitalGusu SchoolNanjing Medical UniversitySuzhouJiangsuChina
| | - Chang‐Hao Xu
- Department of CardiologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
- Cardiovascular Research CenterThe Affiliated Suzhou Hospital of Nanjing Medical UniversitySuzhou Municipal HospitalGusu SchoolNanjing Medical UniversitySuzhouJiangsuChina
| | - Xiang‐Qing Kong
- Department of CardiologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
- Cardiovascular Research CenterThe Affiliated Suzhou Hospital of Nanjing Medical UniversitySuzhou Municipal HospitalGusu SchoolNanjing Medical UniversitySuzhouJiangsuChina
| | - Wei Sun
- Department of CardiologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjingJiangsuChina
- Cardiovascular Research CenterThe Affiliated Suzhou Hospital of Nanjing Medical UniversitySuzhou Municipal HospitalGusu SchoolNanjing Medical UniversitySuzhouJiangsuChina
| |
Collapse
|
23
|
Wuni R, Curi-Quinto K, Liu L, Espinoza D, Aquino AI, Del Valle-Mendoza J, Aguilar-Luis MA, Murray C, Nunes R, Methven L, Lovegrove JA, Penny M, Favara M, Sánchez A, Vimaleswaran KS. Interaction between genetic risk score and dietary carbohydrate intake on high-density lipoprotein cholesterol levels: Findings from the study of obesity, nutrition, genes and social factors (SONGS). Clin Nutr ESPEN 2025; 66:83-92. [PMID: 39800136 DOI: 10.1016/j.clnesp.2024.12.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/18/2024] [Accepted: 12/30/2024] [Indexed: 01/15/2025]
Abstract
BACKGROUND & AIMS Cardiometabolic traits are complex interrelated traits that result from a combination of genetic and lifestyle factors. This study aimed to assess the interaction between genetic variants and dietary macronutrient intake on cardiometabolic traits [body mass index, waist circumference, total cholesterol, high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein cholesterol, triacylglycerol, systolic blood pressure, diastolic blood pressure, fasting serum glucose, fasting serum insulin, and glycated haemoglobin]. METHODS This cross-sectional study consisted of 468 urban young adults aged 20 ± 1 years, and it was conducted as part of the Study of Obesity, Nutrition, Genes and Social factors (SONGS) project, a sub-study of the Young Lives study. Thirty-nine single nucleotide polymorphisms (SNPs) known to be associated with cardiometabolic traits at a genome-wide significance level (P < 5 × 10-8) were used to construct a genetic risk score (GRS). RESULTS There were no significant associations between the GRS and any of the cardiometabolic traits. However, a significant interaction was observed between the GRS and carbohydrate intake on HDL-C concentration (Pinteraction = 0.0007). In the first tertile of carbohydrate intake (≤327 g/day), participants with a high GRS (>37 risk alleles) had a higher concentration of HDL-C than those with a low GRS (≤37 risk alleles) [Beta = 0.06 mmol/L, 95 % confidence interval (CI), 0.01-0.10; P = 0.018]. In the third tertile of carbohydrate intake (>452 g/day), participants with a high GRS had a lower concentration of HDL-C than those with a low GRS (Beta = -0.04 mmol/L, 95 % CI -0.01 to -0.09; P = 0.027). A significant interaction was also observed between the GRS and glycaemic load (GL) on the concentration of HDL-C (Pinteraction = 0.002). For participants with a high GRS, there were lower concentrations of HDL-C across tertiles of GL (Ptrend = 0.017). There was no significant interaction between the GRS and glycaemic index on the concentration of HDL-C, and none of the other GRS∗macronutrient interactions were significant. CONCLUSIONS Our results suggest that young adults who consume a higher carbohydrate diet and have a higher GRS have a lower HDL-C concentration, which in turn is linked to cardiovascular diseases, and indicate that personalised nutrition strategies targeting a reduction in carbohydrate intake might be beneficial for these individuals.
Collapse
Affiliation(s)
- Ramatu Wuni
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, RG6 6DZ, UK.
| | - Katherine Curi-Quinto
- Instituto de Investigación Nutricional (IIN), Av. La Molina 1885, Lima, 15024, Peru.
| | - Litai Liu
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, RG6 6DZ, UK.
| | - Dianela Espinoza
- Group for the Analysis of Development (GRADE), Lima, 15063, Peru.
| | - Anthony I Aquino
- Instituto de Investigación Nutricional (IIN), Av. La Molina 1885, Lima, 15024, Peru
| | - Juana Del Valle-Mendoza
- Instituto de Investigación Nutricional (IIN), Av. La Molina 1885, Lima, 15024, Peru; Biomedicine Laboratory, Research Center of the Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas, Lima, 15087, Peru.
| | - Miguel Angel Aguilar-Luis
- Instituto de Investigación Nutricional (IIN), Av. La Molina 1885, Lima, 15024, Peru; Biomedicine Laboratory, Research Center of the Faculty of Health Sciences, Universidad Peruana de Ciencias Aplicadas, Lima, 15087, Peru.
| | - Claudia Murray
- Department of Real Estate and Planning, University of Reading, Reading, RG6 6UD, UK.
| | - Richard Nunes
- Department of Real Estate and Planning, University of Reading, Reading, RG6 6UD, UK.
| | - Lisa Methven
- Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, RG6 6DZ, UK.
| | - Julie A Lovegrove
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, RG6 6DZ, UK; Institute for Food, Nutrition, and Health (IFNH), University of Reading, Reading, RG6 6AP, UK.
| | - Mary Penny
- Instituto de Investigación Nutricional (IIN), Av. La Molina 1885, Lima, 15024, Peru.
| | - Marta Favara
- Oxford Department of International Development, University of Oxford, Oxford, OX1 3TB, UK.
| | - Alan Sánchez
- Group for the Analysis of Development (GRADE), Lima, 15063, Peru; Oxford Department of International Development, University of Oxford, Oxford, OX1 3TB, UK.
| | - Karani Santhanakrishnan Vimaleswaran
- Hugh Sinclair Unit of Human Nutrition, Department of Food and Nutritional Sciences and Institute for Cardiovascular and Metabolic Research (ICMR), University of Reading, Reading, RG6 6DZ, UK; Institute for Food, Nutrition, and Health (IFNH), University of Reading, Reading, RG6 6AP, UK.
| |
Collapse
|
24
|
El Rouby N, Owusu‐Obeng A, Preuss MH, Lee S, Shi M, Nadukuru R, Van Driest SL, Mosley JD, DelBello M. Genome Wide Association Study (GWAS) Identifies Novel Genetic Loci for Second-Generation Antipsychotics (SGA)-Induced Metabolic Syndrome (MetS). Clin Transl Sci 2025; 18:e70216. [PMID: 40259522 PMCID: PMC12011641 DOI: 10.1111/cts.70216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Accepted: 03/24/2025] [Indexed: 04/23/2025] Open
Abstract
Second-generation antipsychotics (SGA) are widely used for treating psychiatric disorders; however, their use is associated with an increased risk of metabolic syndrome (MetS). To identify common genetic associations of SGA-induced metabolic syndrome (SGA-MetS), we conducted a genome-wide association study (GWAS) in a diverse patient population within the BioVU and BioMe electronic health records (EHRs)-linked biobanks. Additionally, we performed Mendelian Randomization (MR) analysis to investigate the association between the individual metabolic parameters comprising MetS (body mass index [BMI], fasting glucose, blood pressure, HDL, and triglycerides) and SGA-MetS. The meta-analysis of European ancestry GWAS from BioVU and BioMe (N = 9248) identified a genome-wide signal (rs61900075, β = -0.27, SE = 0.05, p = 1.6 × 10-8) on chromosome 11. Multiple associated variants met the suggestive level of association (p ≤ 10-5) in the PELO-ITGA1 locus on chromosome 5 and were associated among the Hispanic Ancestry within BioMe. The meta-analysis of the African Ancestry patients of BioVU and BioMe (N = 2018) identified multiple genome-wide signals that were functionally mapped to NPPC-DIS3L2 in chromosome 2. Finally, the inverse-variance weighted average MR (BMI: OR = 1.2, 95% CI: 1.1-1.4, p = 0.002) showed that genetically predicted, higher BMI was associated with an increased risk of SGA-MetS. Similar results were seen in the sensitivity analyses using the weighted median and Egger MR. This study identified novel variants for SGA-MetS and suggested a role of BMI in increasing the risk of SGA-MetS. The findings highlight the value of EHR biobanks for identifying the genetics underlying SGA-MetS. The associations in chromosome 2 and 5 will need further replication.
Collapse
Affiliation(s)
- Nihal El Rouby
- James L Winkle College of PharmacyUniversity of CincinnatiCincinnatiOHUSA
- Department of Psychiatry & Behavioral NeuroscienceCollege of Medicine, University of CincinnatiCincinnatiOhioUSA
- St. Elizabeth HealthcareEdgewoodKentuckyUSA
| | - Aniwaa Owusu‐Obeng
- The Charles Bronfman Institute for Personalized MedicineIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Michael H. Preuss
- The Charles Bronfman Institute for Personalized MedicineIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Simon Lee
- The Charles Bronfman Institute for Personalized MedicineIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Mingjian Shi
- Department of Biomedical InformaticsVUMCNashvilleTennesseeUSA
| | - Rajiv Nadukuru
- The Charles Bronfman Institute for Personalized MedicineIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Sara L. Van Driest
- Department of PediatricsVanderbilt University Medical Center (VUMC)NashvilleTennesseeUSA
- All of Us Research ProgramNational Institutes of HealthBethesdaMarylandUSA
| | - Jonathan D. Mosley
- Department of Biomedical InformaticsVUMCNashvilleTennesseeUSA
- Department of MedicineVUMCNashvilleTennesseeUSA
| | - Melissa DelBello
- Department of Psychiatry & Behavioral NeuroscienceCollege of Medicine, University of CincinnatiCincinnatiOhioUSA
- College of MedicineUniversity of CincinnatiCincinnatiOHUSA
| |
Collapse
|
25
|
Shi H, Peng X, Lin Y, Song H, Liu L, Zeng Y, He B, Gu Y. Association between different obesity metrics and risk of inguinal hernia. Updates Surg 2025; 77:567-574. [PMID: 39821601 DOI: 10.1007/s13304-025-02062-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: 09/01/2024] [Accepted: 01/07/2025] [Indexed: 01/19/2025]
Abstract
PURPOSE Obesity is closely associated with a lower risk of inguinal hernia, but the association between different obesity metrics and the risk of inguinal hernia is still unclear. METHODS In our study, we categorized obesity measurement indicators into three groups based on the difficulty of measurement: (1) indicators easily available, such as body mass index (BMI), waist circumference (WC), hip circumference (HC), and waist-to-hip ratio (WHR); (2) indicators accessible with moderate difficulty, such as body fat percentage and body fat mass; (3) indicators difficultly accessible, such as the volume of visceral adipose tissue (VAT) and subcutaneous adipose tissue (SAT). Mendelian randomization (MR) analysis was used to investigate the causal relationship between various adiposity measures and the risk of inguinal hernia in both European ancestry and East Asians. RESULTS We identified a total of 17,096 patients with inguinal hernia in the FinnGen cohort and 1664 cases in the Japan Biobank cohort. For European ancestry, MR analysis reported a significant causal association between one standard deviation increase of BMI, WC, HC, body fat percentage, and body fat mass and the lower risk of inguinal hernia, rather than WHR, VAT, and SAT. After the adjustment of BMI, increased WC is still causally associated with a lower risk of inguinal hernia (OR: 0.52; 95% CI: 0.33-0.80; P < 0.01). Among East Asians, only body fat mass is causally associated with a reduced risk of inguinal hernia, rather than BMI, WC, and HC. CONCLUSION Obesity is causally associated with a relatively lower risk of inguinal hernia. The association between different obesity measures and the risk of inguinal hernia has ethnic specificity. These findings help us deepen our understanding of the intrinsic causal relationship between fat distribution and the risk of inguinal hernias at the genetic level.
Collapse
Affiliation(s)
- Hekai Shi
- Department of General Surgery, Huadong Hospital, Fudan University, No. 221, West Yan'an Road, Jing'an District, Shanghai, 200040, People's Republic of China
| | - Xiaoyu Peng
- Department of General Surgery, Huadong Hospital, Fudan University, No. 221, West Yan'an Road, Jing'an District, Shanghai, 200040, People's Republic of China
| | - Yiming Lin
- Department of General Surgery, Huadong Hospital, Fudan University, No. 221, West Yan'an Road, Jing'an District, Shanghai, 200040, People's Republic of China
| | - Heng Song
- Department of General Surgery, Huadong Hospital, Fudan University, No. 221, West Yan'an Road, Jing'an District, Shanghai, 200040, People's Republic of China
| | - Ligang Liu
- Institute of Therapeutic Innovations and Outcomes, College of Pharmacy, The Ohio State University, Columbus, OH, USA
| | - Yihong Zeng
- Department of General Surgery, Huadong Hospital, Fudan University, No. 221, West Yan'an Road, Jing'an District, Shanghai, 200040, People's Republic of China
| | - Binbin He
- Department of General Surgery, Huadong Hospital, Fudan University, No. 221, West Yan'an Road, Jing'an District, Shanghai, 200040, People's Republic of China
| | - Yan Gu
- Department of General Surgery, Huadong Hospital, Fudan University, No. 221, West Yan'an Road, Jing'an District, Shanghai, 200040, People's Republic of China.
| |
Collapse
|
26
|
Wang J, Tian S, Du J, Du S, Chen W, Liu Y. The hypothalamic estrogen receptor α pathway is involved in high-intensity interval training-induced visceral fat loss in premenopausal rats. Lipids Health Dis 2025; 24:118. [PMID: 40148843 PMCID: PMC11948781 DOI: 10.1186/s12944-025-02533-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 03/14/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Visceral adipose tissue (VAT) is strongly associated with metabolic diseases. Both high-intensity interval training (HIT) and moderate-intensity training (MIT) reduce VAT effectively; however, HIT might mediate greater VAT loss in females. The estrogen receptor α (ERα) pathway may play a key role. The aim of the present study was to confirm the role of adipose/hypothalamic ERα in HIT/MIT-mediated VAT loss, as well as the associated hypothalamic electrophysiology and body catabolism changes in pre- and post-menopausal animal models. METHODS Ovariectomy (OVX) or sham surgeries were conducted to establish pre/postmenopausal female rat models. After distance-matched long-term HIT and MIT interventions, ERα expression in hypothalamic/VAT, as well as food intake, spontaneous physical activity (SPA), VAT mass and morphology, local field potential (LFPs) in paraventricular nuclei (PVN) and excessive post-exercise oxygen consumption (EPOC), were observed. A target chemical block during the post-exercise recovery period was executed to further verify the role of the hypothalamic ERα pathway. RESULTS HIT enhanced the expression of ERα in the hypothalamus rather than VAT in the pre-, but not the postmenopausal group, which was accompanied by elevated LFP power density in α and β bands, enhanced EPOC and larger VAT loss than MIT. Chemical blockade of ERα suppressed EPOC and VAT catabolism mediated by HIT. CONCLUSION During the post-exercise recovery period, the hypothalamic ERα pathway involved in HIT induced EPOC elevation and VAT reduction in premenopausal female rats.
Collapse
Affiliation(s)
- Juanjuan Wang
- School of Physical Education, Hebei Normal University, Shijiazhuang, China
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Shuai Tian
- School of Physical Education, Hebei Normal University, Shijiazhuang, China
| | - Jinchan Du
- School of Physical Education, Hebei Normal University, Shijiazhuang, China
| | - Sihao Du
- School of Physical Education, Hebei Normal University, Shijiazhuang, China
| | - Wei Chen
- School of Physical Education, Hebei Normal University, Shijiazhuang, China
- Provincial Key Lab of Measurement and Evaluation in Human Movement and Bio- Information, Hebei Normal University, Shijiazhuang, China
| | - Yang Liu
- School of Physical Education, Hebei Normal University, Shijiazhuang, China.
- Provincial Key Lab of Measurement and Evaluation in Human Movement and Bio- Information, Hebei Normal University, Shijiazhuang, China.
- School of Physical Education, Hebei Normal University, No. 20, South Second Ring Road East, Shijiazhuang, Hebei, China.
| |
Collapse
|
27
|
Ma X, Ding L, Li S, Fan Y, Wang X, Han Y, Yuan H, Sun L, He Q, Liu M. Druggable genome-wide Mendelian randomization identifies therapeutic targets for metabolic dysfunction-associated steatotic liver disease. Lipids Health Dis 2025; 24:113. [PMID: 40140823 PMCID: PMC11938603 DOI: 10.1186/s12944-025-02515-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Metabolic dysfunction-associated steatotic liver disease (MASLD) affects > 25% of the global population, potentially leading to severe hepatic and extrahepatic complications, including metabolic dysfunction-associated steatohepatitis. Given that the pathophysiology of MASLD is incompletely understood, identifying therapeutic targets and optimizing treatment strategies are crucial for addressing this severe condition. METHODS Mendelian randomization (MR) analysis was conducted using two genome-wide association study datasets: a European meta-analysis (8,434 cases; 770,180 controls) and an additional study (3,954 cases; 355,942 controls), identifying therapeutic targets for MASLD. Of 4302 drug-target genes, 2,664 genetic instrument variables were derived from cis-expression quantitative trait loci (cis-eQTLs). Colocalization analyses assessed shared causal variants between MASLD-associated single nucleotide polymorphisms and eQTLs. Using the drug target gene cis-eQTL of liver tissue from the genotype-tissue expression project, we performed MR and summary MR to validate the significance of the gene results of the blood eQTL MR. RNA-sequencing data from liver biopsies were validated using immunohistochemistry and quantitative polymerase chain reaction (qPCR) tests to confirm gene expression findings. RESULT MR analysis across both datasets identified significant MR associations between MASLD and two drug targets-milk fat globule-EGF factor 8 (MFGE8) (odds ratio [OR] 0.89, 95% confidence interval [CI] 0.85-0.94; P = 2.15 × 10-6) and cluster of differentiation 33 (CD33) (OR 1.17, 95% CI 1.10-1.25; P = 1.39 × 10-6). Both targets exhibited strong colocalization with MASLD. Genetic manipulation indicating MFGE8 activation and CD33 inhibition did not increase the risk for other metabolic disorders. RNA-sequencing, qPCR, and immunohistochemistry validation demonstrated consistent differential expressions of MFGE8 and CD33 in MASLD. CONCLUSION CD33 inhibition can reduce MASLD risk, while MFGE8 activation may offer therapeutic benefits for MASLD treatment.
Collapse
Affiliation(s)
- Xiaohui Ma
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
- Department of Endocrinology and Metabolism, Baotou Central Hospital, Baotou, China
| | - Li Ding
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Shuo Li
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Yu Fan
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Xin Wang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Yitong Han
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Hengjie Yuan
- Department of Pharmacy, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| | - Longhao Sun
- Department of General Surgery, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| | - Qing He
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| |
Collapse
|
28
|
Lin X, Liang B, Lam TH, Cheng KK, Zhang W, Xu L. The mediating roles of anthropo-metabolic biomarkers on the association between beverage consumption and breast cancer risk. Nutr J 2025; 24:46. [PMID: 40121496 PMCID: PMC11929343 DOI: 10.1186/s12937-025-01110-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Accepted: 03/02/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Breast cancer (BC) is the most common malignancy in women, yet the role of beverage consumption in BC risk remains unclear. Additionally, the contribution of anthropo-metabolic biomarkers as mediators is unknown, limiting the development of effective prevention strategies. METHODS This study included 13,567 participants from the Guangzhou Biobank Cohort Study (GBCS), where beverage consumption was assessed at baseline using a food frequency questionnaire. BC cases were identified through cancer registry linkage over a mean follow-up of 14.8 years. Mendelian randomization (MR) analyses were performed to evaluate the causal effects of beverage consumption on BC risk, with a two-step MR approach used to estimate mediation effects. RESULTS During follow-up, 243 BC cases were identified. Weekly consumption of ≥ 1 portion of sugar sweetened beverages (SSB), versus < 1 portion, was significantly associated with a higher risk of BC (hazard ratio [HR] 1.58, 95% confidence interval [CI] 1.12-2.23). This association was partly mediated by body mass index (proportion mediated [PM] 4.2%, 95% CI 0.9-17.1%) and uric acid (PM 18.8%, 95% CI 1.5-77.5%). Weekly consumption of > 6 portions of dairy-based milk was associated with a non-significantly higher BC risk (HR 1.41, 95% CI 0.99-2.03), while 3-6 portions of soy milk were associated with a lower BC risk (HR 0.31, 95% CI 0.10-0.98). No significant associations were found for pure fruit juice, coffee, tea, or alcoholic drinks. MR analyses supported the detrimental effect of SSB on BC risk, with high-density lipoprotein cholesterol, polyunsaturated fatty acids to total fatty acids (TFAs) ratio, and omega-6 fatty acids to TFAs ratio mediating 2.44%, 2.73%, and 3.53% of the association, respectively. CONCLUSION This study suggested that SSB consumption was a risk factor for BC and identified key anthropo-metabolic biomarkers mediating this relationship. Reducing SSB consumption and addressing associated metabolic pathways may offer effective strategies for BC prevention.
Collapse
Affiliation(s)
- Xiaoyi Lin
- School of Public Health, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, Guangdong Province, China
- Greater Bay Area, Greater Bay Area Public Health Research Collaboration, Guangzhou, China
| | - Boheng Liang
- Guangzhou Center for Disease Control and Prevention, Guangzhou, China
| | - Tai Hing Lam
- School of Public Health, the University of Hong Kong, Hong Kong, China
- Guangzhou Twelfth People's Hospital, Guangzhou, China
- Greater Bay Area, Greater Bay Area Public Health Research Collaboration, Guangzhou, China
| | - Kar Keung Cheng
- School of Health Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK
| | - Weisen Zhang
- Guangzhou Twelfth People's Hospital, Guangzhou, China
- Greater Bay Area, Greater Bay Area Public Health Research Collaboration, Guangzhou, China
| | - Lin Xu
- School of Public Health, Sun Yat-sen University, No. 74 Zhongshan 2nd Road, Guangzhou, Guangdong Province, China.
- School of Public Health, the University of Hong Kong, Hong Kong, China.
- School of Health Sciences, College of Medicine and Health, University of Birmingham, Birmingham, UK.
- Greater Bay Area, Greater Bay Area Public Health Research Collaboration, Guangzhou, China.
| |
Collapse
|
29
|
Chen XX, Lu FY, Wang Y, Zhang L, Li SQ, Lin YN, Yan YR, Ding YJ, Li N, Zhou JP, Sun XW, Li QY. Causal effect of life-course adiposity on the risk of respiratory diseases: a Mendelian randomization study. Nutr Metab (Lond) 2025; 22:25. [PMID: 40119483 PMCID: PMC11929217 DOI: 10.1186/s12986-025-00915-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 03/03/2025] [Indexed: 03/24/2025] Open
Abstract
BACKGROUND There is limited evidence on the causal associations of life-course adiposity with the risk of respiratory diseases. This study aimed to elucidate these associations. METHODS Two-sample Mendelian randomization was conducted using genetic instruments of life-course adiposity (including birth weight, childhood BMI, and adulthood adiposity) to estimate their causal effect on respiratory diseases in participants of European ancestry from the UK Biobank, the FinnGen consortium, and other large consortia. RESULTS Genetically predicted higher birth weight was associated with decreased risk of acute upper respiratory infections and increased risk of pulmonary embolism, sleep apnea, and lung cancer. Genetically predicted high childhood BMI was associated with increased risk of asthma, COPD, pulmonary embolism, and sleep apnea. However, most of these observed associations were no longer significant after adjusting for adult BMI. Genetically predicted higher adult BMI and WHR were associated with 10 and 4 respiratory diseases, respectively. High adult body fat percentage and visceral adiposity were genetically associated with increased risk of 9 and 11 respiratory diseases, respectively. Consistently, genetically predicted higher whole-body fat mass was associated with increased risk of 8 respiratory diseases. CONCLUSIONS This study provides genetic evidence that greater adiposity in childhood and adulthood has a causal effect in increasing the risk of a wide range of respiratory diseases. Furthermore, the effects of childhood obesity on respiratory outcomes may be mediated by adult obesity.
Collapse
Affiliation(s)
- Xi Xi Chen
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Fang Ying Lu
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yi Wang
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Liu Zhang
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shi Qi Li
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ying Ni Lin
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ya Ru Yan
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yong Jie Ding
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ning Li
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jian Ping Zhou
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xian Wen Sun
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Qing Yun Li
- Department of Respiratory and Critical Care Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- College of Health Science and Technology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Institute of Respiratory Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
30
|
Wang Z, Chen L, Kang R, Li Z, Fan J, Peng Y, He Y, Zhao X. Mendelian Randomization Analysis Identifies Causal Effects of Multi-Site Chronic Pain on Obstructive Sleep Apnea. Nat Sci Sleep 2025; 17:463-473. [PMID: 40124580 PMCID: PMC11929529 DOI: 10.2147/nss.s487056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 02/11/2025] [Indexed: 03/25/2025] Open
Abstract
Background Observational studies have suggested an association between obstructive sleep apnea (OSA) and chronic pain disorders, but causal evidence have not been confirmed. Methods Here we performed Mendelian randomization (MR) study to explore the potential causal association and mediating roles of modifiable factors between multi-site chronic pain (MCP) and OSA. Independent single nucleotide polymorphisms (SNPs) (N=26) from MCP GWAS (n=387,649) in the UK Biobank were used as instrumental variables to test associations with OSA from the FinnGen consortium, which encompassed 16,761 individuals with OSA cases and 201,194 individuals without OSA. Results MR analyses provide genetic evidence to predict MCP on the risk of OSA. Specifically, a per-site increase in multi-site chronic pain was linked to a 184% higher risk of OSA (ORIVW = 1.84, 95% CI = 1.29-2.63, p = 7.24×10-4). However, we also performed reverse association analyses and found no significant casual effect of OSA on MCP. MR estimates were in agreement regardless of the method used, such as MR-egger, weighted median and weighted mode, thereby demonstrating the accuracy of the causal associations. Through mediation analyses, we found that body mass index (BMI), waist circumference, and educational attainment explained a substantial proportion of the association between MCP and OSA (proportion mediated=21.13%; 26.57% and 9.66% respectively). Conclusion Our findings suggest that both pain management interventions, prevention of obesity and health education are likely to be effective strategies to reduce OSA risk in individuals with MCP.
Collapse
Affiliation(s)
- Zuxing Wang
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu, 610072, People’s Republic of China
| | - Lili Chen
- Sichuan Provincial Center for Mental Health, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Key Laboratory of Psychosomatic Medicine, Chinese Academy of Medical Sciences, Chengdu, 610072, People’s Republic of China
| | - Ruishi Kang
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Zhuowei Li
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Jiangang Fan
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Yi Peng
- Department of Otorhinolaryngology Head and Neck Surgery, Second People’s Hospital of Chengdu, Chengdu, 610000, People’s Republic of China
| | - Yunqi He
- Sichuan Provincial Key Laboratory for Human Disease Gene Study, Center for Medical Genetics and Department of Laboratory Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, People’s Republic of China
- Research Unit for Blindness Prevention, Chinese Academy of Medical Sciences (2019RU026), Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital, Chengdu, Sichuan, People’s Republic of China
| | - Xiaolong Zhao
- Department of Otolaryngology Head and Neck Surgery, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| |
Collapse
|
31
|
Lee Y, Seo JH, Lee J, Kim HS. Causal Effects of 25-Hydroxyvitamin D on Metabolic Syndrome and Metabolic Risk Traits: A Bidirectional Two-Sample Mendelian Randomization Study. Biomedicines 2025; 13:723. [PMID: 40149699 PMCID: PMC11940704 DOI: 10.3390/biomedicines13030723] [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: 01/24/2025] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Individuals with metabolic syndrome (MetS) present reduced 25(OH)D levels. We performed a two-sample Mendelian randomization (MR) study to investigate whether causal relationships exist between 25(OH)D levels and MetS/MetS risk traits, including waist circumference, body mass index (BMI), hypertension (systolic/diastolic blood pressure), triglyceride, high-density lipoprotein cholesterol, and glucose levels. Methods: We employed genetic variants related to 25(OH)D levels from the SUNLIGHT Consortium and a European genome-wide association study meta-analysis, including UK Biobank (UKB) data, as well as variants for MetS and MetS risk traits from UKB and multiple European consortia. Several MR methods were used, i.e., inverse-variance weighted, weighted median, and MR-Egger regression. Heterogeneity and horizontal pleiotropy analyses were performed to ensure the stability of candidate single-nucleotide polymorphisms (SNPs) as the instrumental variable. We first conducted univariable MR to investigate the relationship between 25(OH)D levels and MetS, including its related risk traits, and subsequently performed multivariable MR to adjust for potential confounders. Results: This study did not provide evidence of a causal relationship between 25(OH)D levels and MetS/MetS risk traits. However, we found that several risk traits of MetS, such as waist circumference, BMI, and TG, had an inverse-causal relationship with 25(OH)D levels, suggesting that 25(OH)D levels could be secondary consequences of metabolic illnesses. Conclusions: We identified no causal relationship between 25(OH)D levels and MetS/MetS risk factors. However, 25(OH)D levels may result from MetS traits.
Collapse
Affiliation(s)
- Young Lee
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul 05368, Republic of Korea; (Y.L.); (J.H.S.)
| | - Je Hyun Seo
- Veterans Medical Research Institute, Veterans Health Service Medical Center, Seoul 05368, Republic of Korea; (Y.L.); (J.H.S.)
| | - Junyong Lee
- Department of Family Medicine, Veterans Health Service Medical Center, Seoul 05368, Republic of Korea;
| | - Hwa Sun Kim
- Department of Family Medicine, Veterans Health Service Medical Center, Seoul 05368, Republic of Korea;
| |
Collapse
|
32
|
Baya NA, Erdem IS, Venkatesh SS, Reibe S, Charles PD, Navarro-Guerrero E, Hill B, Lassen FH, Claussnitzer M, Palmer DS, Lindgren CM. Combining evidence from human genetic and functional screens to identify pathways altering obesity and fat distribution. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.09.19.24313913. [PMID: 39371160 PMCID: PMC11451655 DOI: 10.1101/2024.09.19.24313913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Overall adiposity and body fat distribution are heritable traits associated with altered risk of cardiometabolic disease and mortality. Performing rare variant (minor allele frequency<1%) association testing using exome-sequencing data from 402,375 participants in the UK Biobank (UKB) for nine overall and tissue-specific fat distribution traits, we identified 19 genes where putatively damaging rare variation associated with at least one trait (Bonferroni-adjusted P <1.58×10 -7 ) and 50 additional genes at FDR≤1% ( P ≤4.37×10 -5 ). These 69 genes exhibited significantly higher (one-sided t -test P =3.58×10 -18 ) common variant prioritisation scores than genes not significantly enriched for rare putatively damaging variation, with evidence of monotonic allelic series (dose-response relationships) among ultra-rare variants (minor allele count≤10) in 22 genes. Combining rare and common variation evidence, allelic series and longitudinal analysis, we selected 14 genes for CRISPR knockdown in human white adipose tissue cell lines. In three previously uncharacterised target genes, knockdown increased (two-sided t -test P <0.05) lipid accumulation, a cellular phenotype relevant for fat mass traits, compared to Cas9-empty negative controls: COL5A3 (fold change [FC]=1.72, P =0.0028), EXOC7 (FC=1.35, P =0.0096), and TRIP10 (FC=1.39, P =0.0157); furthermore, knockdown of PPARG (FC=0.25, P =5.52×10 -7 ) and SLTM (FC=0.51, P =1.91×10 -4 ) resulted in reduced lipid accumulation. Integrating across population-based genetic and in vitro functional evidence, we highlight therapeutic avenues for altering obesity and body fat distribution by modulating lipid accumulation.
Collapse
|
33
|
Chen R, Petrazzini BO, Duffy Á, Rocheleau G, Jordan D, Bansal M, Do R. Trans-ancestral rare variant association study with machine learning-based phenotyping for metabolic dysfunction-associated steatotic liver disease. Genome Biol 2025; 26:50. [PMID: 40065360 PMCID: PMC11892324 DOI: 10.1186/s13059-025-03518-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Genome-wide association studies (GWAS) have identified common variants associated with metabolic dysfunction-associated steatotic liver disease (MASLD). However, rare coding variant studies have been limited by phenotyping challenges and small sample sizes. We test associations of rare and ultra-rare coding variants with proton density fat fraction (PDFF) and MASLD case-control status in 736,010 participants of diverse ancestries from the UK Biobank, All of Us, and BioMe and performed a trans-ancestral meta-analysis. We then developed models to accurately predict PDFF and MASLD status in the UK Biobank and tested associations with these predicted phenotypes to increase statistical power. RESULTS The trans-ancestral meta-analysis with PDFF and MASLD case-control status identifies two single variants and two gene-level associations in APOB, CDH5, MYCBP2, and XAB2. Association testing with predicted phenotypes, which replicates more known genetic variants from GWAS than true phenotypes, identifies 16 single variants and 11 gene-level associations implicating 23 additional genes. Two variants were polymorphic only among African ancestry participants and several associations showed significant heterogeneity in ancestry and sex-stratified analyses. In total, we identified 27 genes, of which 3 are monogenic causes of steatosis (APOB, G6PC1, PPARG), 4 were previously associated with MASLD (APOB, APOC3, INSR, PPARG), and 23 had supporting clinical, experimental, and/or genetic evidence. CONCLUSIONS Our results suggest that trans-ancestral association analyses can identify ancestry-specific rare and ultra-rare coding variants in MASLD pathogenesis. Furthermore, we demonstrate the utility of machine learning in genetic investigations of difficult-to-phenotype diseases in trans-ancestral biobanks.
Collapse
Affiliation(s)
- Robert Chen
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ben Omega Petrazzini
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Genomic Data Analytics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Áine Duffy
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Genomic Data Analytics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ghislain Rocheleau
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Genomic Data Analytics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel Jordan
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Genomic Data Analytics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Meena Bansal
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ron Do
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Center for Genomic Data Analytics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
34
|
Fu S, Li Q, Cheng L, Wan S, Wang Q, Min Y, Xie Y, Liu H, Hu T, Liu H, Chen W, Zhang Y, Xiong F. Causal Relationship Between Intelligence, Noncognitive Education, Cognition and Urinary Tract or Kidney Infection: A Mendelian Randomization Study. Int J Nephrol Renovasc Dis 2025; 18:71-85. [PMID: 40070673 PMCID: PMC11895678 DOI: 10.2147/ijnrd.s511736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/25/2025] [Indexed: 03/14/2025] Open
Abstract
Background The occurrence of urinary tract or kidney infection is correlated with intelligence, noncognitive education and cognition, but the causal relationship between them remains uncertain, and which risk factors mediate this causal relationship remains unknown. Methods The intelligence (n=269,867), noncognitive education (n=510,795) and cognition data (n=257,700) were obtained from genome-wide association studies (GWAS) conducted in individuals of European ethnicities. The genetic associations between these factors and urinary tract or kidney infection (UK Biobank, n=397,867) were analyzed using linkage disequilibrium score regression. We employed a two-sample univariate and multivariate Mendelian randomization to evaluate the causal relationship, and utilized a two-step Mendelian randomization to examine the involvement of 28 potential mediators and their respective mediating proportions. Results The genetic correlation coefficients of intelligence, noncognitive education, cognition, and urinary tract or kidney infection were -0.338, -0.218, and -0.330. The Mendelian randomization using the inverse variance weighted method revealed each 1-SD increase in intelligence, the risk of infection decreased by 15.9%, while after adjusting for noncognitive education, the risk decreased by 20%. For each 1-SD increase in noncognitive education, the risk of infection decreased by 8%, which further reduced to 7.1% after adjusting for intelligence and to 6.7% after adjusting for cognition. For each 1-SD increase in cognition, the risk of infection decreased by 10.8%, increasing to 11.9% after adjusting for noncognitive education. The effects of intelligence and cognition are interdependent. 2 out of 28 potential mediating factors exhibited significant mediation effects in the causal relationship between noncognitive education and urinary tract or kidney infection, with body mass index accounting for 12.1% of the mediation effect and smoking initiation accounting for 14.7%. Conclusion Enhancing intelligence, noncognitive education, and cognition can mitigate the susceptibility to urinary tract or kidney infection. Noncognitive education exhibited independent effect, while body mass index and smoking initiation assuming a mediating role.
Collapse
Affiliation(s)
- Shuai Fu
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Qiang Li
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Li Cheng
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Sheng Wan
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Quan Wang
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Yonglong Min
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Yanghao Xie
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Huizhen Liu
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Taotao Hu
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Hong Liu
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Weidong Chen
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Yanmin Zhang
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| | - Fei Xiong
- Department of Nephrology, Wuhan No. 1 Hospital, Wuhan, Hubei Province, People’s Republic of China
| |
Collapse
|
35
|
Mokhtari A, Ibrahim EC, Gloaguen A, Barrot CC, Cohen D, Derouin M, Vachon H, Charbonnier G, Loriod B, Decraene C, Yalcin I, Marie-Claire C, Etain B, Belzeaux R, Delahaye-Duriez A, Lutz PE. Using multiomic integration to improve blood biomarkers of major depressive disorder: a case-control study. EBioMedicine 2025; 113:105569. [PMID: 39914267 PMCID: PMC11848217 DOI: 10.1016/j.ebiom.2025.105569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 12/12/2024] [Accepted: 01/14/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Major depressive disorder (MDD) is a leading cause of disability, with a twofold increase in prevalence in women compared to men. Over the last few years, identifying molecular biomarkers of MDD has proven challenging, reflecting interactions among multiple environmental and genetic factors. Recently, epigenetic processes have been proposed as mediators of such interactions, with the potential for biomarker development. METHODS We characterised gene expression and two mechanisms of epigenomic regulation, DNA methylation (DNAm) and microRNAs (miRNAs), in blood samples from a cohort of individuals with MDD and healthy controls (n = 169). Case-control comparisons were conducted for each omic layer. We also defined gene coexpression networks, followed by step-by-step annotations across omic layers. Third, we implemented an advanced multiomic integration strategy, with covariate correction and feature selection embedded in a cross-validation procedure. Performance of MDD prediction was systematically compared across 6 methods for dimensionality reduction, and for every combination of 1, 2 or 3 types of molecular data. Feature stability was further assessed by bootstrapping. FINDINGS Results showed that molecular and coexpression changes associated with MDD were highly sex-specific and that the performance of MDD prediction was greater when the female and male cohorts were analysed separately, rather than combined. Importantly, they also demonstrated that performance progressively increased with the number of molecular datasets considered. INTERPRETATION Informational gain from multiomic integration had already been documented in other medical fields. Our results pave the way toward similar advances in molecular psychiatry, and have practical implications for developing clinically useful MDD biomarkers. FUNDING This work was supported by the Centre National de la Recherche Scientifique (contract UPR3212), the University of Strasbourg, the Université Sorbonne Paris Nord, the Université Paris Cité, the Fondation de France (FdF N° Engt:00081244 and 00148126; ECI, IY, RB, PEL), the French National Research Agency (ANR-18-CE37-0002, BE, CMC, ADD, PEL, ECI; ANR-18-CE17-0009, ADD; ANR-19-CE37-0010, PEL; ANR-21-RHUS-009, ADD, BE, CMC, CCB; ANR-22-PESN-0013, ADD), the Fondation pour la Recherche sur le Cerveau (FRC 2019, PEL), Fondation de France (2018, BE, CMC, ADD) and American Foundation for Suicide Prevention (AFSP YIG-1-102-19; PEL).
Collapse
Affiliation(s)
- Amazigh Mokhtari
- Université Paris Cité, Inserm, NeuroDiderot, UMR-1141, 75019, Paris, France
| | - El Chérif Ibrahim
- Aix-Marseille Univ, CNRS, INT, Inst Neurosci Timone, 13005, Marseille, France
| | - Arnaud Gloaguen
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, 91000, Evry, France
| | | | - David Cohen
- Université Paris Cité, Inserm, NeuroDiderot, UMR-1141, 75019, Paris, France
| | - Margot Derouin
- Université Paris Cité, Inserm, NeuroDiderot, UMR-1141, 75019, Paris, France
| | - Hortense Vachon
- Aix-Marseille Université, INSERM, TAGC, 13009, Marseille, France
| | | | - Béatrice Loriod
- Aix-Marseille Université, INSERM, TAGC, 13009, Marseille, France
| | - Charles Decraene
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives UPR 3212, F-67000, Strasbourg, France
| | - Ipek Yalcin
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives UPR 3212, F-67000, Strasbourg, France; Department of Psychiatry and Neuroscience, Université Laval, Québec, QC, G1V 0A6, Canada
| | - Cynthia Marie-Claire
- Université Paris Cité, INSERM UMR-S 1144, Optimisation thérapeutique en neuropsychopharmacologie, OTeN, F-75006, Paris, France
| | - Bruno Etain
- Université Paris Cité, INSERM UMR-S 1144, Optimisation thérapeutique en neuropsychopharmacologie, OTeN, F-75006, Paris, France; Assistance Publique des Hôpitaux de Paris, GHU Lariboisière-Saint Louis-Fernand Widal, DMU Neurosciences, Département de psychiatrie et de Médecine Addictologique, F-75010, Paris, France
| | - Raoul Belzeaux
- Aix-Marseille Univ, CNRS, INT, Inst Neurosci Timone, 13005, Marseille, France; Département de psychiatrie, CHU de Montpellier, Montpellier, France
| | - Andrée Delahaye-Duriez
- Université Paris Cité, Inserm, NeuroDiderot, UMR-1141, 75019, Paris, France; Unité fonctionnelle de médecine génomique et génétique clinique, Hôpital Jean Verdier, Assistance Publique des Hôpitaux de Paris, F-93140, Bondy, France; Université Sorbonne Paris Nord, F-93000, Bobigny, France.
| | - Pierre-Eric Lutz
- Centre National de la Recherche Scientifique, Université de Strasbourg, Institut des Neurosciences Cellulaires et Intégratives UPR 3212, F-67000, Strasbourg, France; Douglas Mental Health University Institute, McGill University, QC, H4H 1R3, Montréal, Canada.
| |
Collapse
|
36
|
Higa R, Pourteymour S, Kolan PS, Dankel SN, Fernø J, Mellgren G, Pan C, Seldin MM, Lusis AJ, Drevon CA, Dalen KT, Norheim FA. Hepatic lipid metabolism is altered in Ubiad1 +/- mice of both sexes. Sci Rep 2025; 15:7022. [PMID: 40016272 PMCID: PMC11868635 DOI: 10.1038/s41598-025-91283-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 02/19/2025] [Indexed: 03/01/2025] Open
Abstract
UbiA prenyltransferase domain containing 1 (Ubiad1) has the potential to affect cholesterol and phospholipid levels in different cell types. We previously identified Ubiad1 as a candidate gene for regulating subcutaneous fat pad weight in a mouse genome-wide association study. Here we evaluated the relationship between Ubiad1 and obesity-related traits in cohorts of humans and mice, and in Ubiad1+/- mice fed a high-fat diet. In both humans and mice, adipose tissue Ubiad1 mRNA expression correlated negatively with adiposity and positively with mitochondria-related genes. To determine the role of Ubiad1 in high-fat diet-induced obesity, we disrupted the Ubiad1 gene in mice. Deletion of Ubiad1 was embryonically lethal in C57BL/6 N mice, preventing analysis of adult Ubiad1-/- mice. Thus, male and female Ubiad1+/+ and Ubiad1+/- mice were fed high-fat diet for 10 weeks, with no difference in weight gain and adipose tissue organ weights observed between the genotypes. Analysis of liver mRNA expression revealed that Ubiad1 heterozygosis (Ubiad1+/-) altered several pathways involved in lipid metabolism. Detailed lipid quantification with HPLC-qTOF/MS showed increased levels of hepatic ceramides in female Ubiad1+/- mice, whereas phosphatidylglycerols, phosohatidylinositol and lysophosphatidylethanolamines were reduced in male Ubiad1+/- mice. Our findings reveal sex-specific effects of Ubiad1 expression on hepatic lipid metabolism.
Collapse
Affiliation(s)
- Ryoko Higa
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Sognsvannsveien 9, Domus Medica, 0372 Oslo, Norway
- Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
| | - Shirin Pourteymour
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Sognsvannsveien 9, Domus Medica, 0372 Oslo, Norway
| | - Pratibha S Kolan
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Sognsvannsveien 9, Domus Medica, 0372 Oslo, Norway
| | - Simon N Dankel
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Johan Fernø
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Gunnar Mellgren
- Hormone Laboratory, Department of Medical Biochemistry and Pharmacology, Haukeland University Hospital, Bergen, Norway
- Mohn Center for Diabetes Precision Medicine, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Calvin Pan
- Department of Medicine, Division of Cardiology, University of California, 650 Charles E Young Drive South, Los Angeles, Los Angeles, CA, 90095, USA
- Departments of Human Genetics & Microbiology, Immunology, and Molecular Genetics, University of California, los Angeles, 650 Charles E. Young Drive South, Los Angeles, CA, 90095, USA
| | - Marcus M Seldin
- Department of Biological Chemistry, University of California, Irvine, Irvine, USA
| | - Aldons J Lusis
- Department of Medicine, Division of Cardiology, University of California, 650 Charles E Young Drive South, Los Angeles, Los Angeles, CA, 90095, USA
- Departments of Human Genetics & Microbiology, Immunology, and Molecular Genetics, University of California, los Angeles, 650 Charles E. Young Drive South, Los Angeles, CA, 90095, USA
| | - Christian A Drevon
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Sognsvannsveien 9, Domus Medica, 0372 Oslo, Norway
- Vitas AS, Science Park, Gaustadalléen 21, 0349, Oslo, Norge
| | - Knut T Dalen
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Sognsvannsveien 9, Domus Medica, 0372 Oslo, Norway
| | - Frode A Norheim
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Sognsvannsveien 9, Domus Medica, 0372 Oslo, Norway.
| |
Collapse
|
37
|
Li H, Yin G, Zhang Y, Wang Z, Lv F, Li R, Qin J, Ye X. Effect of fat distribution on left ventricular structure and function in different sexes: a Mendelian randomization study. Front Endocrinol (Lausanne) 2025; 16:1355968. [PMID: 40070582 PMCID: PMC11893391 DOI: 10.3389/fendo.2025.1355968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 02/07/2025] [Indexed: 03/14/2025] Open
Abstract
Background Although there is an interaction between sex, body fat distribution, and cardiac structure and function, these relationships have not been fully elucidated yet. This study aims to reveal the causal relationship between genetic determinants of fat distribution pattern and function of the left ventricular structure in different sexes. Methods Genetic variants for waist circumference, hip circumference, waist-to-hip ratio (WHR), and body mass index (BMI) were selected from genome-wide association studies conducted in European samples. The dataset for left ventricular (LV) parameters was obtained from over 35,000 European samples in the UK Biobank Cardiovascular Magnetic Resonance sub-study. Two-sample Mendelian randomization (MR) analysis was employed to explore causal relationships. Results After adjusting for BMI, WHR shows a positive causal relationship with LV hypertrophy and a significant negative causal relationship with LV volume and diastolic function. In further subgroup analysis, we only found similar results in WHR among the female population (FWHR), while in the male population, there was no significant causal relationship between MWHR and LV hypertrophy and diastolic function. Additionally, in our MR analysis, no causal relationship was found between WHR and LVEF. Conclusions This study indicates that the fat distribution pattern has unique effects on the structure and function of the LV, and these effects vary by sex. This study provides evidence for a causal relationship between fat distribution and LV structure and function across both sexes.
Collapse
Affiliation(s)
- Hang Li
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Guangjiao Yin
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Zhongnan Hospital of Wuhan University, Institute of Hepatobiliary Diseases of Wuhan University, Transplant Center of Wuhan University, Wuhan, Hubei, China
| | - Yanfang Zhang
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ziwei Wang
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Fang Lv
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Rui Li
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Juanjuan Qin
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Center for Healthy Aging, Wuhan University School of Nursing, Wuhan, Hubei, China
| | - Xujun Ye
- Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
- Center for Healthy Aging, Wuhan University School of Nursing, Wuhan, Hubei, China
| |
Collapse
|
38
|
Loh NY, Vasan SK, Rosoff DB, Roberts E, van Dam AD, Verma M, Phillips D, Wesolowska-Andersen A, Neville MJ, Noordam R, Ray DW, Tobias JH, Gregson CL, Karpe F, Christodoulides C. LRP5 promotes adipose progenitor cell fitness and adipocyte insulin sensitivity. COMMUNICATIONS MEDICINE 2025; 5:51. [PMID: 40000740 PMCID: PMC11862225 DOI: 10.1038/s43856-025-00774-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND WNT signaling plays a key role in postnatal bone formation. Individuals with gain-of-function mutations in the WNT co-receptor LRP5 exhibit increased lower-body fat mass and potentially enhanced glucose metabolism, alongside high bone mass. However, the mechanisms by which LRP5 regulates fat distribution and its effects on systemic metabolism remain unclear. This study aims to explore the role of LRP5 in adipose tissue biology and its impact on metabolism. METHODS Metabolic assessments and imaging were conducted on individuals with gain- and loss-of-function LRP5 mutations, along with age- and BMI-matched controls. Mendelian randomization analyses were used to investigate the relationship between bone, fat distribution, and systemic metabolism. Functional studies and RNA sequencing were performed on abdominal and gluteal adipose cells with LRP5 knockdown. RESULTS Here we show that LRP5 promotes lower-body fat distribution and enhances systemic and adipocyte insulin sensitivity through cell-autonomous mechanisms, independent of its bone-related functions. LRP5 supports adipose progenitor cell function by activating WNT/β-catenin signaling and preserving valosin-containing protein (VCP)-mediated proteostasis. LRP5 expression in adipose progenitors declines with age, but gain-of-function LRP5 variants protect against age-related fat loss in the lower body. CONCLUSIONS Our findings underscore the critical role of LRP5 in regulating lower-body fat distribution and insulin sensitivity, independent of its effects on bone. Pharmacological activation of LRP5 in adipose tissue may offer a promising strategy to prevent age-related fat redistribution and metabolic disorders.
Collapse
Affiliation(s)
- Nellie Y Loh
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Senthil K Vasan
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Daniel B Rosoff
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Emile Roberts
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Andrea D van Dam
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Manu Verma
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Daniel Phillips
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Agata Wesolowska-Andersen
- Nuffield Department of Medicine, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Matt J Neville
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, OUH Foundation Trust, Oxford, UK
| | - Raymond Noordam
- Department of Internal Medicine, Section of Gerontology and Geriatrics, Leiden University Medical Center, Leiden, The Netherlands
| | - David W Ray
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, OUH Foundation Trust, Oxford, UK
| | - Jonathan H Tobias
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, Southmead Hospital, University of Bristol, Bristol, UK
| | - Celia L Gregson
- MRC Integrative Epidemiology Unit (IEU), Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, Southmead Hospital, University of Bristol, Bristol, UK
| | - Fredrik Karpe
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, OUH Foundation Trust, Oxford, UK
| | - Constantinos Christodoulides
- Radcliffe Department of Medicine, Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK.
- NIHR Oxford Biomedical Research Centre, OUH Foundation Trust, Oxford, UK.
| |
Collapse
|
39
|
Welch BM, Bommarito PA, Cantonwine DE, Milne GL, Stevens DR, Edin ML, Zeldin DC, Meeker JD, McElrath TF, Ferguson KK. Consumer Product Chemical Mixtures and Oxylipin-Mediated Inflammation and Oxidative Stress during Early Pregnancy: Findings from a Large US Pregnancy Cohort. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:2987-2999. [PMID: 39913660 DOI: 10.1021/acs.est.4c10390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/19/2025]
Abstract
Consumer product chemicals pose an environmental risk to public health. Exposure during pregnancy to consumer product chemicals, particularly phthalates and phenols, may increase the susceptibility to pregnancy disorders by dysregulating inflammation and oxidative stress. However, existing studies rely on downstream and nonmodifiable markers of these processes. Oxylipins are oxidized lipids that act as key upstream drivers of inflammation and oxidative stress. Importantly, oxylipins are responsive to therapeutic interventions and thus potentially modifiable. Using recent advances in lipidomics and statistical approaches to address both individual chemical biomarkers and their mixtures, we determined associations between early pregnancy biomarkers of consumer product chemical exposure and oxylipins in a large prospective cohort. Overall, our results revealed associations among oxylipins produced across several biosynthetic pathways, suggesting a pattern indicative of dysregulated inflammation and elevated levels of oxidative stress. Phthalate metabolites were the primary drivers of associations, particularly for metabolites of low molecular weight phthalates, often used in personal care products. However, we found similar associations for a biomarker of a phthalate replacement that is increasingly used in consumer products. Our study provides observational evidence of specific physiological pathways that may be dysregulated by exposure to consumer product chemicals, including legacy phthalates and phthalate replacements.
Collapse
Affiliation(s)
- Barrett M Welch
- School of Public Health, University of Nevada, 1664 N. Virginia Street, Reno, Nevada 89557, United States
- Epidemiology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, 111 TW Alexander Dr, Durham, North Carolina 27709, United States
| | - Paige A Bommarito
- Epidemiology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, 111 TW Alexander Dr, Durham, North Carolina 27709, United States
| | - David E Cantonwine
- Division of Maternal-Fetal Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Ginger L Milne
- Department of Medicine, Vanderbilt School of Medicine, Nashville, Tennessee 37232, United States
| | - Danielle R Stevens
- Epidemiology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, 111 TW Alexander Dr, Durham, North Carolina 27709, United States
| | - Matthew L Edin
- Immunity, Inflammation, and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, 111 TW Alexander Dr, Durham, North Carolina 27709, United States
| | - Darryl C Zeldin
- Immunity, Inflammation, and Disease Laboratory, Division of Intramural Research, National Institute of Environmental Health Sciences, 111 TW Alexander Dr, Durham, North Carolina 27709, United States
| | - John D Meeker
- Department of Environmental Health Sciences, University of Michigan School of Public Health, 6635 SPH Tower, 109 S. Observatory Street, Ann Arbor, Michigan 48109, United States
| | - Thomas F McElrath
- Department of Medicine, Vanderbilt School of Medicine, Nashville, Tennessee 37232, United States
| | - Kelly K Ferguson
- Epidemiology Branch, Division of Intramural Research, National Institute of Environmental Health Sciences, 111 TW Alexander Dr, Durham, North Carolina 27709, United States
| |
Collapse
|
40
|
Yuan W, Xu X, Zhang X, Fan W, Zhou W, Zhao F. Exploring the Associations of Obesity and Glycemic Traits with Retinal Vein Occlusion: A Univariate and Multivariable Mendelian Randomization Study. Ophthalmic Epidemiol 2025:1-9. [PMID: 39919303 DOI: 10.1080/09286586.2025.2458245] [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: 07/17/2024] [Revised: 11/24/2024] [Accepted: 01/19/2025] [Indexed: 02/09/2025]
Abstract
PURPOSE To explore the genetic links between obesity, glycemic traits and retinal vein occlusion (RVO). METHODS Summary-level statistics for obesity and glycemic traits were extracted from publicly available genome-wide association studies (GWAS) of European participants in the IEU Open GWAS database. Genetic associations with clinically diagnosed RVO were obtained from the FinnGenresearch project (372 cases and 182,573 controls). Two-sample Mendelian randomization (MR) and multivariate MR (MVMR) analysis were performed to determine the total effect and direct effect, respectively. RESULTS After adjustment for the false discovery rate (FDR), the primary inverse-variance-weighted (IVW) methods indicated that the odds ratios of RVO increased with per 1-standard deviation increased in body mass index (BMI) (OR = 1.94, 95% CI: 1.23-3.08,p-FDR = 0.025), waist circumference (OR = 2.4, 95% CI: 1.36-4.24, p-FDR = 0.019), fasting glucose (OR = 5.01, 95% CI: 2-12.55, p-FDR = 0.0067) and two-hour glucose (OR = 3.17, 95% CI: 1.63-6.18,p-FDR = 0.0067). Higher whole-body fat-free mass (OR = 0.45, 95% CI: 0.26-0.8,p-FDR = 0.025) is a potential protective factor for RVO. In addition, the results of MVMR showed that BMI, whole-body fat-free mass, fasting glucose and two-hour glucose were independent factors that had a direct impact on the onset of RVO. CONCLUSIONS Our comprehensive MR analysis suggested significant genetic associations between BMI, whole-body fat-free mass, fasting glucose, two-hour glucose and RVO. This study highlighted the importance of weight, blood glucose management and physical activity for primary prevention and control of RVO.
Collapse
Affiliation(s)
- Weichen Yuan
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
- Department of Ophthalmology, Key Lens Research Laboratory of Liaoning Province, Shenyang, China
| | - Xin Xu
- Department of Biochemistry and Molecular Biology, China Medical University, Shenyang, China
| | - Xiran Zhang
- Department of Optometry, China Medical University, Shenyang, China
| | - Wenqi Fan
- Department of Optometry, China Medical University, Shenyang, China
| | - Wenkai Zhou
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
- Department of Ophthalmology, Key Lens Research Laboratory of Liaoning Province, Shenyang, China
| | - Fangkun Zhao
- Department of Ophthalmology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
- Department of Ophthalmology, Key Lens Research Laboratory of Liaoning Province, Shenyang, China
| |
Collapse
|
41
|
Fu L, Liu Q, Cheng H, Zhao X, Xiong J, Mi J. Insights Into Causal Effects of Genetically Proxied Lipids and Lipid-Modifying Drug Targets on Cardiometabolic Diseases. J Am Heart Assoc 2025; 14:e038857. [PMID: 39868518 PMCID: PMC12074789 DOI: 10.1161/jaha.124.038857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/13/2024] [Indexed: 01/28/2025]
Abstract
BACKGROUND The differential impact of serum lipids and their targets for lipid modification on cardiometabolic disease risk is debated. This study used Mendelian randomization to investigate the causal relationships and underlying mechanisms. METHODS Genetic variants related to lipid profiles and targets for lipid modification were sourced from the Global Lipids Genetics Consortium. Summary data for 10 cardiometabolic diseases were compiled from both discovery and replication data sets. Expression quantitative trait loci data from relevant tissues were employed to evaluate significant lipid-modifying drug targets. Comprehensive analyses including colocalization, mediation, and bioinformatics were conducted to validate the results and investigate potential mediators and mechanisms. RESULTS Significant causal associations were identified between lipids, lipid-modifying drug targets, and various cardiometabolic diseases. Notably, genetic enhancement of LPL (lipoprotein lipase) was linked to reduced risks of myocardial infarction (odds ratio [OR]1, 0.65 [95% CI, 0.57-0.75], P1=2.60×10-9; OR2, 0.59 [95% CI, 0.49-0.72], P2=1.52×10-7), ischemic heart disease (OR1, 0.968 [95% CI, 0.962-0.975], P1=5.50×10-23; OR2, 0.64 [95% CI, 0.55-0.73], P2=1.72×10-10), and coronary heart disease (OR1, 0.980 [95% CI, 0.975-0.985], P1=3.63×10-14; OR2, 0.64 [95% CI, 0.54-0.75], P2=6.62×10-8) across 2 data sets. Moreover, significant Mendelian randomization and strong colocalization associations for the expression of LPL in blood and subcutaneous adipose tissue were linked with myocardial infarction (OR, 0.918 [95% CI, 0.872-0.967], P=1.24×10-3; PP.H4, 0.99) and coronary heart disease (OR, 0.991 [95% CI, 0.983-0.999], P=0.041; PP.H4=0.92). Glucose levels and blood pressure were identified as mediators in the total effect of LPL on cardiometabolic outcomes. CONCLUSIONS The study substantiates the causal role of lipids in specific cardiometabolic diseases, highlighting LPL as a potent drug target. The effects of LPL are suggested to be influenced by changes in glucose and blood pressure, providing insights into its mechanism of action.
Collapse
Affiliation(s)
- Liwan Fu
- Center for Non‐Communicable Disease ManagementBeijing Children’s Hospital, Capital Medical University, National Center for Children’s HealthBeijingChina
| | - Qin Liu
- Department of UltrasoundChildren’s Hospital of the Capital Institute of PediatricsBeijingChina
| | - Hong Cheng
- Department of EpidemiologyCapital Institute of PediatricsBeijingChina
| | - Xiaoyuan Zhao
- Department of EpidemiologyCapital Institute of PediatricsBeijingChina
| | - Jingfan Xiong
- Child and Adolescent Chronic Disease Prevention and Control DepartmentShenzhen Center for Chronic Disease ControlShenzhenChina
| | - Jie Mi
- Center for Non‐Communicable Disease ManagementBeijing Children’s Hospital, Capital Medical University, National Center for Children’s HealthBeijingChina
- Key Laboratory of Major Diseases in Children, Ministry of EducationChina
| |
Collapse
|
42
|
Liu F, Wang K, Nie J, Deng MG. Unraveling the Link: Mendelian Randomization Reveals Causal Relationship Between Selenium and Metabolic Syndrome. Biol Trace Elem Res 2025; 203:790-798. [PMID: 38776021 DOI: 10.1007/s12011-024-04237-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 05/15/2024] [Indexed: 11/28/2024]
Abstract
Observational studies have linked selenium and metabolic syndrome (MetS), but the causality remains unclear. Therefore, this study intends to determine the causal relationship between selenium and the risk of MetS and its component features [body mass index (BMI), waist circumference adjusted for BMI (WCadjBMI), triglycerides (TC), HDL-cholesterol (HDL-C), fasting blood glucose (FBG), fasting blood insulin (FBI), systolic blood pressure (SBP), and diastolic blood pressure (DBP)]. This study was designed as the two-sample Mendelian randomization (MR), and genetic variants were obtained from the genome-wide association studies. The inverse variance weighted (IVW) was applied as the primary method, and the MR-Egger, weighted median, and MR-PRESSO were supplemented to assess its robustness. The Bonferroni method was used to correct p-values for multiple tests. Genetically incremented selenium level was related to higher odds ratios of developing the MetS (OR = 1.054, 95% CI = 1.016-1.094, p = 0.0049). As for components, significant causal links were identified between selenium and BMI (β = 0.015, p = 1.321 × 10-5), WCadjBMI (β = 0.033, p = 2.352 × 10-4), HDL-C (β = -0.036, p = 1.352 × 10-8), FBG (β = 0.028, p = 0.001), and FBI (β = 0.028, p = 0.002). No significant association was discovered for SBP (β = -0.076, p = 0.218) and DBP (β = 0.054, p = 0.227). These results were generally supported by the weighted median and MR-PRESSO methods. Our study provided evidence of the causal effect of selenium on MetS risk from the genetic perspective in the European population, and further investigation across diverse populations was warranted.
Collapse
Affiliation(s)
- Fang Liu
- School of Public Health, Wuhan University, Wuhan, 430071, China
| | - Kai Wang
- Department of Public Health, Wuhan Fourth Hospital, Wuhan, 430000, China
| | - Jiaqi Nie
- Xiaogan Municipal Center for Disease Control and Prevention, Xiaogan, 432000, Hubei, China
| | - Ming-Gang Deng
- Department of Psychiatry, Wuhan Mental Health Center, Wuhan, 430012, Hubei, China.
- Department of Psychiatry, Wuhan Hospital for Psychotherapy, Wuhan, 430012, Hubei, China.
| |
Collapse
|
43
|
Miller MA. Time for bed: diet, sleep and obesity in children and adults. Proc Nutr Soc 2025; 84:45-52. [PMID: 38012858 DOI: 10.1017/s0029665123004846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Sufficient sleep is necessary for optimal health, daytime performance and wellbeing and the amount required is age-dependent and decreases across the lifespan. Sleep duration is usually affected by age and several different cultural, social, psychological, behavioural, pathophysiological and environmental factors. This review considers how much sleep children and adults need, why this is important, what the consequences are of insufficient sleep and how we can improve sleep. A lack of the recommended amount of sleep for a given age group has been shown to be associated with detrimental effects on health including effects on metabolism, endocrine function, immune function and haemostatic pathways. Obesity has increased worldwide in the last few decades and the WHO has now declared it a global epidemic. A lack of sleep is associated with an increased risk of obesity in children and adults, which may lead to future poor health outcomes. Data from studies in both children and adults suggest that the relationship between sleep and obesity may be mediated by several different mechanisms including alterations in appetite and satiety, sleep timing, circadian rhythm and energy balance. Moreover, there is evidence to suggest that improvements in sleep, in both children and adults, can be beneficial for weight management and diet and certain foods might be important to promote sleep. In conclusion this review demonstrates that there is a wide body of evidence to suggest that sleep and obesity are causally related and recommends that further research is required to inform policy, and societal change.
Collapse
|
44
|
Ghosh S, Bouchard C. Considerations on efforts needed to improve our understanding of the genetics of obesity. Int J Obes (Lond) 2025; 49:206-210. [PMID: 38849463 PMCID: PMC11805711 DOI: 10.1038/s41366-024-01528-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 06/09/2024]
Affiliation(s)
- Sujoy Ghosh
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA.
| | - Claude Bouchard
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA.
| |
Collapse
|
45
|
Yang LZ, Yang Y, Hong C, Wu QZ, Shi XJ, Liu YL, Chen GZ. Systematic Mendelian Randomization Exploring Druggable Genes for Hemorrhagic Strokes. Mol Neurobiol 2025; 62:1359-1372. [PMID: 38977622 PMCID: PMC11772512 DOI: 10.1007/s12035-024-04336-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 06/26/2024] [Indexed: 07/10/2024]
Abstract
Patients with hemorrhagic stroke have high rates of morbidity and mortality, and drugs for prevention are very limited. Mendelian randomization (MR) analysis can increase the success rate of drug development by providing genetic evidence. Previous MR analyses only analyzed the role of individual drug target genes in hemorrhagic stroke; therefore, we used MR analysis to systematically explore the druggable genes for hemorrhagic stroke. We sequentially performed summary-data-based MR analysis and two-sample MR analysis to assess the associations of all genes within the database with intracranial aneurysm, intracerebral hemorrhage, and their subtypes. Validated genes were further analyzed by colocalization. Only genes that were positive in all three analyses and were druggable were considered desirable genes. We also explored the mediators of genes affecting hemorrhagic stroke incidence. Finally, the associations of druggable genes with other cardiovascular diseases were analyzed to assess potential side effects. We identified 56 genes that significantly affected hemorrhagic stroke incidence. Moreover, TNFSF12, SLC22A4, SPARC, KL, RELT, and ADORA3 were found to be druggable. The inhibition of TNFSF12, SLC22A4, and SPARC can reduce the risk of intracranial aneurysm, subarachnoid hemorrhage, and intracerebral hemorrhage. Gene-induced hypertension may be a potential mechanism by which these genes cause hemorrhagic stroke. We also found that blocking these genes may cause side effects, such as ischemic stroke and its subtypes. Our study revealed that six druggable genes were associated with hemorrhagic stroke, and the inhibition of TNFSF12, SLC22A4, and SPARC had preventive effects against hemorrhagic strokes.
Collapse
Affiliation(s)
- Lun-Zhe Yang
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yong Yang
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Chuan Hong
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Qi-Zhe Wu
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xiong-Jie Shi
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yi-Lin Liu
- Department of Neurosurgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Guang-Zhong Chen
- Department of Neurosurgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| |
Collapse
|
46
|
Hu M, Li X, Wu J, Li B, Xia J, Yang Y, Yin C. Phenome-Wide Investigation of the Causal Associations Between Pre-Pregnancy Obesity Traits and Gestational Diabetes: A Two-Sample Mendelian Randomization Analyses. Reprod Sci 2025; 32:395-403. [PMID: 38789873 DOI: 10.1007/s43032-024-01577-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/19/2024] [Indexed: 05/26/2024]
Abstract
Pre-pregnancy obesity was associated with gestational diabetes in observational studies, but whether this relationship is causal remains to be determined. To evaluate whether pre-pregnancy obesity traits causally affect gestational diabetes risk, a two-sample Mendelian randomization (MR) analysis was performed utilizing summary-level statistics from published genome-wide association studies (GWAS). Obesity-related traits included body mass index (BMI), overweight, obesity, obesity class 1, obesity class 2, obesity class 3, childhood obesity, waist circumference (WC), hip circumference (HC), waist-to-hip ratio (WHR), percent liver fat, visceral adipose tissue volume, abdominal subcutaneous adipose tissue volume. Effect estimates were evaluated using the inverse-variance weighting method. Weighted median, MR-Egger, simple mode, and weighted mode were performed as sensitivity analyses. Genetically predicted pre-pregnancy BMI [odds ratio (OR) = 1.68; 95% confidence interval (CI): 1.45-1.95; P = 9.13 × 10-12], overweight (OR = 1.49; 95% CI: 1.21-1.85; P = 2.06 × 10-4), obesity (OR = 1.25; 95% CI: 1.18-1.33; P = 8.01 × 10-13), obesity class 1 (OR = 1.31; 95% CI: 1.17-1.46; P = 1.49 × 10-6), obesity class 2 (OR = 1.26; 95% CI: 1.16-1.37; P = 5.23 × 10-8), childhood obesity (OR = 1.33; 95% CI: 1.23-1.44; P = 4.06 × 10-12), and WHR (OR = 2.35; 95% CI: 1.44-3.83; P = 5.89 × 10-4) were associated with increased risk of gestational diabetes. No significant association was observed with obesity class 3, WC, HC, percent liver fat, visceral adipose tissue volume, or abdominal subcutaneous adipose tissue volume. Similar results were observed in sensitivity analyses. Therefore, genetically predicted pre-pregnancy obesity traits may increase the risk of gestational diabetes. Weight control before pregnancy may be beneficial to prevent gestational diabetes.
Collapse
Affiliation(s)
- Mengjin Hu
- Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Xiaosong Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China
| | - Jiangong Wu
- Fenyang Center for Disease Control and Prevention, Fenyang, 032200, Shanxi Province, China
| | - Boyu Li
- Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Jinggang Xia
- Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yuejin Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100037, China.
| | - Chunlin Yin
- Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
47
|
Morales-Berstein F, Khouja J, Gormley M, Ebrahimi E, Virani S, McKay J, Brennan P, Richardson TG, Relton CL, Smith GD, Borges MC, Dudding T, Richmond RC. Reassessing the link between adiposity and head and neck cancer: a Mendelian randomization study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2024.11.21.24317707. [PMID: 39974030 PMCID: PMC11838947 DOI: 10.1101/2024.11.21.24317707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Background Adiposity has been associated with an increased risk of head and neck cancer (HNC). Although body mass index (BMI) has been inversely associated with HNC risk among smokers, this is likely due to confounding. Previous Mendelian randomization (MR) studies could not fully discount causality between adiposity and HNC due to limited statistical power. Hence, we aimed to revisit this using the largest genome-wide association study (GWAS) of HNC available, which has more granular data on HNC subsites. Methods We assessed the genetically predicted effects of BMI (N=806,834), waist-to-hip ratio (WHR; N=697,734) and waist circumference (N=462,166) on the risk of HNC (N=12,264 cases and 19,259 controls) and its subsites (oral, laryngeal, hypopharyngeal and oropharyngeal cancers) using a two-sample MR framework. We used the inverse variance weighted (IVW) MR approach and multiple sensitivity analyses including the weighted median, weighted mode, MR-Egger, MR-PRESSO, and CAUSE approaches. We also used multivariable MR (MVMR) to explore the direct effects of the adiposity measures on HNC, while accounting for smoking behaviour, a well-known HNC risk factor. Results In univariable MR, higher genetically predicted BMI increased the risk of overall HNC (IVW OR=1.17 per 1 standard deviation [1-SD] higher BMI, 95% CI 1.02-1.34, p=0.03), with no heterogeneity across subsites (Q p=0.78). However, the effect was not consistent in sensitivity analyses. The IVW effect was attenuated when smoking was included in the MVMR model (OR accounting for comprehensive smoking index=0.96 per 1-SD higher BMI, 95% CI 0.80-1.15, p=0.64) and CAUSE indicated the IVW results could be biased by correlated pleiotropy. Furthermore, we did not find a link between genetically predicted WHR (IVW OR=1.05 per 1-SD higher WHR, 95% CI 0.89-1.24, p=0.53) or waist circumference and HNC risk (IVW OR=1.01 per 1-SD higher waist circumference, 95% CI 0.85-1.21, p=0.87). Conclusions Our findings suggest that adiposity does not play a role in HNC risk.
Collapse
Affiliation(s)
- Fernanda Morales-Berstein
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Jasmine Khouja
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Mark Gormley
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- University of Bristol Dental School, 1 Trinity Walk, Avon Street, Bristol, United Kingdom
| | - Elmira Ebrahimi
- Genomic Epidemiology Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Shama Virani
- Genomic Epidemiology Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - James McKay
- Genomic Epidemiology Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Paul Brennan
- Genomic Epidemiology Branch, International Agency for Research on Cancer, World Health Organization, Lyon, France
| | - Tom G Richardson
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Caroline L Relton
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- London School of Hygiene & Tropical Medicine, Keppel Street, London WC1E 7HT, United Kingdom
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - M Carolina Borges
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Tom Dudding
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- University of Bristol Dental School, 1 Trinity Walk, Avon Street, Bristol, United Kingdom
| | - Rebecca C Richmond
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
48
|
Mollahosseini M, Yazdanpanah Z, Nadjarzadeh A, Mirzaei M, Kalantar SM, Ashrafzadeh HR, Mirzaei K, Mozaffari-Khosravi H. Interactions between DASH-style diet and ADIPOQ and ARL15 genes polymorphisms on blood pressure and central obesity in Iranian adults. Sci Rep 2025; 15:3298. [PMID: 39865127 PMCID: PMC11770118 DOI: 10.1038/s41598-025-87567-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 01/20/2025] [Indexed: 01/28/2025] Open
Abstract
This study aimed to investigate the interactions between the Dietary approaches to stop hypertension (DASH) and the rs1501299 and rs6450176 SNPs in terms of cardiometabolic risk factors. This cross-sectional study was conducted on 387 adults (20-70 years old) residing in Yazd, Iran. The participants were selected from participants in the recruitment phase of the Yazd Health Study (YaHS) which is a population-based cohort of 9,962 adults. The genotypes of rs1501299 and rs6450176 SNPs were determined using the restriction fragment length polymorphism (RFLP) method. Unlike for the G allele carriers of rs1501299, high adherence to the DASH dietary pattern was related to the lower diastolic blood pressure (DBP; P-interaction = 0.014) and systolic blood pressure (SBP; P-interaction = 0.051) in the participants with TT genotype. Moreover, in individuals with the AA genotype of rs6450176 SNP, adherence to this diet was associated with lower SBP (P-interaction = 0.012), waist-to-hip ratio (WHR; P-interaction = 0.026) and waist-to-height ratio (WHtR; P-interaction = 0.040), while in G allele carriers, the mean of these items was greater in individuals with high adherence to the DASH diet. The results suggest that following the DASH diet may be related to lower blood pressure and central obesity indices only in individuals with TT genotype of rs1501299 and AA genotype of rs6450176, respectively, whereas it can be associated with higher WHR and WHtR in G allele carriers of rs6450176.
Collapse
Affiliation(s)
- Mehdi Mollahosseini
- Molecular and Medicine Research Center, Khomein University of Medical Sciences, Khomein, Iran
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Zeinab Yazdanpanah
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Azadeh Nadjarzadeh
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Masoud Mirzaei
- Yazd Non-Communicable Diseases Research Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyed Mehdi Kalantar
- Department of Medical Genetics, Shahid Sadoughi University of Medical Science, Yazd, Iran
- Research and Clinical Center for Infertility, Reproduction Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hamid Reza Ashrafzadeh
- Abortion Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Khadijeh Mirzaei
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Hassan Mozaffari-Khosravi
- Nutrition and Food Security Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
- Department of Nutrition, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
- Department of Nutrition, School of Health, Shahid Sadoughi University of Medical Sciences, Shohadaye Gomnam Blvd, Alem square, Yazd, 8915173160, Iran.
| |
Collapse
|
49
|
Guo L, Pan Y, Yang Y, Kong X, Song S, Li M, Zhao Y, Ma X, Wang X, Sun Q. Association of novel metabolic status with asymptomatic intracranial arterial stenosis: A cross-sectional study. Int J Obes (Lond) 2025:10.1038/s41366-025-01723-7. [PMID: 39856246 DOI: 10.1038/s41366-025-01723-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 01/05/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025]
Abstract
OBJECTIVES To investigate the association of metabolic status newly defined or obesity with asymptomatic intracranial arterial stenosis (aICAS) among populations in rural China. METHODS The cross-sectional study is based on the Rose asymptomatic IntraCranial Artery Stenosis (RICAS) cohort, which enrolled 2005 participants aged 40 years or older without a history of clinical stroke or transient ischemic attack. Metabolically healthy status (MH) was defined by a newly proposed criterion: (1) systolic blood pressure (SBP) < 130 mmHg and without antihypertensive medication; (2) a waist-to-hip ratio (WHR) below 1.03 for men and below 0.95 for women; (3) no diabetes. All participants were categorized based on their metabolic status and obesity. Multivariate logistic regression models were used to investigate the association between metabolic status or obesity and aICAS. RESULTS Among 2005 participants, 1597 (79.65%) were defined as metabolically unhealthy status (MU) according to the new criterion. MU was significantly associated with aICAS (OR 2.02, 95% CI 1.11-3.68, P = 0.021), especially moderate-to-severe aICAS (OR 2.43, 95% CI 1.04-5.72, P = 0.042). The prevalence of aICAS increased with the numbers of metabolic disorders (P for linear trend <0.001). Both metabolically unhealthy normal-weight (MUN) (OR 2.11, 95% CI 1.10-4.03, P = 0.025) and metabolically unhealthy obesity (MUO) (OR 3.30, 95% CI 1.64-6.64, P = 0.001) were significantly correlated with aICAS, but not metabolically healthy obesity (MHO). Subgroup analysis further confirmed the association between MU and aICAS risk only in men (P for interaction = 0.042). CONCLUSIONS MU defined by the new criterion was significantly associated with aICAS, especially with moderate-to-severe aICAS. This novel criterion effectively identifies individuals with a high prevalence of aICAS among populations with obesity, which could be crucial for stroke prevention.
Collapse
Affiliation(s)
- Liying Guo
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Yongli Pan
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yumeng Yang
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Xianglong Kong
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Shiqing Song
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
| | - Maoyu Li
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yuanyuan Zhao
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xiaotong Ma
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xiang Wang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Qinjian Sun
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China.
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
50
|
Cely I, Blencowe M, Shu L, Diamante G, Ahn IS, Zhang G, LaGuardia J, Liu R, Saleem Z, Wang S, Davis R, Lusis AJ, Yang X. Glo1 reduction in mice results in age- and sex-dependent metabolic dysfunction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.24.634754. [PMID: 39896461 PMCID: PMC11785252 DOI: 10.1101/2025.01.24.634754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Objectives Advanced glycation end products (AGEs) have been implicated as an important mediator of metabolic disorders including obesity, insulin resistance, and coronary artery disease. Glyoxalase 1 (Glo1) is a critical enzyme in the clearance of toxic dicarbonyl such as methylglyoxal, precursors of AGEs. The role of AGE-independent mechanisms that underly Glo1-induced metabolic disorders have yet to be elucidated. Methods We performed a longitudinal study of female and male Glo1 heterozygous knockdown (Glo1 +/- ) mice with ~50% gene expression and screened metabolic phenotypes such as body weight, adiposity, glycemic control and plasma lipids. We also evaluated atherosclerotic burden, AGE levels, and gene expression profiles across cardiometabolic tissues (liver, adipose, muscle, kidney and aorta) to identify pathway perturbations and potential regulatory genes of Glo1 actions. Results Partial loss of Glo1 resulted in obesity, hyperglycemia, dyslipidemia, and alterations in lipid metabolism in metabolic tissues in an age- and sex-dependent manner. Glo1 +/- females displayed altered glycemic control and increased plasma triglycerides, which aligned with significant perturbations in genes involved in adipogenesis, PPARg, insulin signaling, and fatty acid metabolism pathways in liver and adipose tissues. Conversely, Glo1 +/- males developed increased skeletal muscle mass and visceral adipose depots along with changes in lipid metabolism pathways. For both cohorts, most phenotypes manifested after 14 weeks of age. Evaluation of methylglyoxal-derived AGEs demonstrated changes in only male skeletal muscle but not in female tissues, which cannot explain the broad metabolic changes observed in Glo1 +/- mice. Transcriptional profiles suggest that altered glucose and lipid metabolism may be partially explained by alternative detoxification of methylglyoxal to metabolites such as pyruvate. Moreover, transcription factor (TF) analysis of the tissue-specific gene expression data identified TFs involved in cardiometabolic diseases such as Hnf4a (all tissues) and Arntl (aorta, liver, and kidney) which are female-biased regulators and whose targets are altered in response to Glo1 +/- . Conclusions Our results indicate that Glo1 reduction perturbs metabolic health and metabolic pathways in a sex- and age-dependent manner without significant changes in AGEs across metabolic tissues. Rather, tissue-specific gene expression analysis suggests that key transcription factors such as Hfn4a and Arntl as well as metabolite changes from alternative methylglyoxal detoxification such as pyruvate, likely contribute to metabolic dysregulation in Glo1 +/- mice.
Collapse
Affiliation(s)
- Ingrid Cely
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
- Molecular Toxicology Interdepartmental Program, University of California Los Angeles, Los Angeles, CA, USA
| | - Montgomery Blencowe
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
- Molecular, Cellular, and Integrative Physiology Interdepartmental Ph.D. Program, University of California, Los Angeles, Los Angeles, CA, United States of America
| | - Le Shu
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
- Molecular, Cellular, and Integrative Physiology Interdepartmental Ph.D. Program, University of California, Los Angeles, Los Angeles, CA, United States of America
| | - Graciel Diamante
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - In Sook Ahn
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Guanglin Zhang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jonnby LaGuardia
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ruoshui Liu
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zara Saleem
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Susanna Wang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Richard Davis
- Departments of Medicine, Human Genetics, and Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Aldons J. Lusis
- Departments of Medicine, Human Genetics, and Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Medicine, Human Genetics, and Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, USA
| | - Xia Yang
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, USA
- Molecular, Cellular, and Integrative Physiology Interdepartmental Ph.D. Program, University of California, Los Angeles, Los Angeles, CA, United States of America
- Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, USA
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA
| |
Collapse
|