1
|
Cui Y, Chen Y, Hu M, Zhou H, Guo J, Wang Q, Xu Z, Chen L, Zhang W, Tang S. Bidirectional Mendelian randomization and colocalization analysis of gut microbiota on lipid profile. Comput Biol Chem 2025; 117:108422. [PMID: 40080991 DOI: 10.1016/j.compbiolchem.2025.108422] [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: 02/14/2024] [Revised: 01/03/2025] [Accepted: 03/05/2025] [Indexed: 03/15/2025]
Abstract
The gut microbiota plays a crucial role in human health, but its impact on lipid metabolism remains unclear. Understanding the causal relationship between gut bacteria and lipid profiles is essential for developing strategies to prevent and treat dyslipidemia and cardiovascular diseases. This study aimed to assess this relationship using two-sample Mendelian randomization (MR). Data for both exposure and outcomes were obtained from the IEU-GWAS database, with lipid profile data sourced from a publication. Genome-wide significant single nucleotide polymorphisms (SNPs), which were independent of outcome factors but correlated with exposure variables, were identified as instrumental variables. Several MR methods, including weighted analysis, maximum likelihood, inverse variance weighting (IVW), MR-Egger, and weighted median, were applied. Colocalization analysis further validated the findings. The analysis revealed microbial groups with causal relationships to ApoA1, ApoB, high-density lipoprotein cholesterol, low-density lipoprotein cholesterol, total cholesterol, and triglycerides. Reverse MR and colocalization analysis provided additional confirmation of these results. This study offers new evidence of the causal link between gut microbiota and lipid profiles, providing insights for improving lipid profiles and reducing cardiovascular disease risk.
Collapse
Affiliation(s)
- Yu Cui
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China; Plastic Surgery Institute of Shantou University Medical College, Shantou, Guangdong 515051, China; Shantou Plastic Surgery Clinical Research Center, Shantou, Guangdong 515051, China
| | - Yanzhu Chen
- Operating Room 1 Area, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China
| | - Mengting Hu
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China; Plastic Surgery Institute of Shantou University Medical College, Shantou, Guangdong 515051, China; Shantou Plastic Surgery Clinical Research Center, Shantou, Guangdong 515051, China
| | - He Zhou
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China
| | - Jiarui Guo
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China
| | - Qijia Wang
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China
| | - Zaihua Xu
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China
| | - Liyun Chen
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China; Research Center of Translational Medicine, Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515051, China
| | - Wancong Zhang
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China; Plastic Surgery Institute of Shantou University Medical College, Shantou, Guangdong 515051, China; Shantou Plastic Surgery Clinical Research Center, Shantou, Guangdong 515051, China.
| | - Shijie Tang
- Department of Plastic Surgery and Burns Center, Second Affiliated Hospital, Shantou University Medical College, Shantou, Guangdong 515051, China; Plastic Surgery Institute of Shantou University Medical College, Shantou, Guangdong 515051, China; Shantou Plastic Surgery Clinical Research Center, Shantou, Guangdong 515051, China.
| |
Collapse
|
2
|
Ma H, Wang Y, Yang Y, Chen J, Jin X. Deciphering the shared genetic architecture between bipolar disorder and body mass index. J Affect Disord 2025; 379:127-135. [PMID: 40056998 DOI: 10.1016/j.jad.2025.03.002] [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: 12/19/2024] [Revised: 02/27/2025] [Accepted: 03/01/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND The comorbidity between bipolar disorder (BD) and high body mass index (BMI) is well-documented, but their shared genetic architecture remains unclear. Our study aimed to explore this genetic correlation and potential causality. METHODS Utilizing large-scale genome-wide association study (GWAS) summary statistics, we quantified global and local genetic correlations between BD and BMI using linkage disequilibrium score regression (LDSC) and Heritability Estimation from Summary Statistics. Stratified LDSC characterized genetic overlap across functional categories. Cross-trait meta-analyses identified shared risk single nucleotide polymorphisms (SNPs), followed by colocalization analysis using Coloc. Bi-directional Mendelian randomization (MR) assessed causality, while tissue-level SNP heritability enrichment for BD and BMI was evaluated using LDSC-specific expressed genes and Multi-marker Analysis of Genomic Annotation. RESULTS We found a genetic correlation between BD and BMI, especially in localized genomic regions. Cross-trait meta-analysis identified 46 significant SNPs shared between BD and BMI, including three novel shared risk SNPs. Colocalization analysis verified two novel SNPs with shared causal variants linked to ITIH1 and TM6SF2 genes. MR analysis demonstrated a causal effect of BD on BMI, but not the reverse. Gene expression data revealed genetic correlation enrichment in five specific brain regions. CONCLUSION This study comprehensively analyzes the genetic correlation between BD and BMI, uncovering shared genetic architecture and identifying novel risk loci. These findings provide new insights into the interplay between BD and BMI, informing the development of diagnostic tools and therapeutic strategies.
Collapse
Affiliation(s)
- Haochuan Ma
- Department of Oncology, the Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, Guangdong, China; Guangdong Provincial Hospital of Chinese Medicine Postdoctoral Research Workstation, Guangzhou, Guangdong, China
| | - Yongbin Wang
- Department of Laboratory Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yang Yang
- Department of Laboratory Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Jing Chen
- Department of Laboratory Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China
| | - Xing Jin
- Department of Laboratory Medicine, The Affiliated Hospital of Yangzhou University, Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
3
|
Hawton K, Shirodkar D, Siese T, Hamilton-Shield JP, Giri D. A recent update on childhood obesity: aetiology, treatment and complications. J Pediatr Endocrinol Metab 2025; 38:429-441. [PMID: 40105362 DOI: 10.1515/jpem-2024-0316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 02/22/2025] [Indexed: 03/20/2025]
Abstract
Obesity is a complex, chronic condition characterised by excess adiposity. Rates of obesity in childhood and adolescence are increasing worldwide, with a corresponding increase in adulthood. The aetiology of obesity is multifactorial and results from a combination of endocrine, genetic, environmental and societal factors. Population level approaches to reduce the prevalence of childhood obesity worldwide are urgently needed. There are wide-ranging complications from excess weight affecting every system in the body, which lead to significant morbidity and reduced life expectancy. Treatment of obesity and its complications requires a multi-faceted, biopsychosocial approach incorporating dietary, exercise and psychological treatments. Pharmacological treatments for treating childhood obesity have recently become available, and there is further development of new anti-obesity medications in the pipeline. In addition, bariatric surgery is being increasingly recognised as a treatment option for obesity in adolescence providing the potential to reverse complications related to excess weight. In this review, we present an update on the prevalence, aetiology, complications and treatment of childhood obesity.
Collapse
Affiliation(s)
- Katherine Hawton
- 156596 Bristol Royal Hospital for Children, University Hospitals Bristol and Weston NHS Foundation Trust , Bristol, UK
- University of Bristol, Bristol, UK
| | - Diksha Shirodkar
- 156596 Bristol Royal Hospital for Children, University Hospitals Bristol and Weston NHS Foundation Trust , Bristol, UK
- University of Bristol, Bristol, UK
| | | | - Julian P Hamilton-Shield
- 156596 Bristol Royal Hospital for Children, University Hospitals Bristol and Weston NHS Foundation Trust , Bristol, UK
- NIHR Biomedical Research Centre (Nutrition Theme), University Hospitals Bristol and Weston NHS Foundations Trust, Bristol, UK
| | - Dinesh Giri
- 156596 Bristol Royal Hospital for Children, University Hospitals Bristol and Weston NHS Foundation Trust , Bristol, UK
- University of Bristol, Bristol, UK
| |
Collapse
|
4
|
Masip G, Han HY, Meng T, Nielsen DE. Polygenic Risk and Nutrient Intake Interactions on Obesity Outcomes: A Systematic Review and Meta-Analysis of Observational Studies. Obes Rev 2025:e13941. [PMID: 40375759 DOI: 10.1111/obr.13941] [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: 02/12/2024] [Revised: 03/26/2025] [Accepted: 04/30/2025] [Indexed: 05/18/2025]
Abstract
BACKGROUND Diet is an important determinant of body weight and may modulate genetic susceptibility to obesity. OBJECTIVE This systematic review and meta-analysis aimed to synthesize evidence related to interactions between polygenic risk and nutrient intakes on obesity outcomes. METHODS MEDLINE, EMBASE, Web of Science, and Cochrane Library were systematically searched to identify observational studies that assessed interactions between polygenic risk and nutrient intakes on obesity-related outcomes. Random effects meta-analyses were performed for pooled polygenic risk score (PRS)-total fat intake and PRS-protein intake interaction coefficients on body mass index (BMI). RESULTS Twenty-six publications were retrieved with studies conducted among European, Asian, and African samples. Dietary fats (saturated fat, omega-3, and trans fat) and energy intake were most frequently reported to interact with PRS on obesity outcomes, but the total number of studies available was low. No significant interactions were identified in meta-analyses of PRS interactions with total fat intake and protein intake on BMI. Several studies were rated as low quality, heterogeneity was high, and although study samples were racially diverse, PRSs tended to be based on samples of European ancestry. CONCLUSION Evidence of interactions between polygenic risk and nutrient intakes on obesity outcomes is limited and inconsistent. Further research addressing limitations related to study quality and polygenic risk characterization is needed.
Collapse
Affiliation(s)
- Guiomar Masip
- School of Human Nutrition, McGill University, Montreal, Quebec, Canada
- Growth, Exercise, Nutrition and Development (GENUD), Research Group, Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria Aragón (IIS Aragón) Universidad de Zaragoza, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y la Nutrición (CIBERObn), Instituto de Salud Carlos III, Madrid, Spain
| | - Hannah Yang Han
- School of Human Nutrition, McGill University, Montreal, Quebec, Canada
| | - Tongzhu Meng
- School of Human Nutrition, McGill University, Montreal, Quebec, Canada
| | - Daiva E Nielsen
- School of Human Nutrition, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
5
|
Lin M, Guo J, Tao H, Gu Z, Tang W, Zhou F, Jiang Y, Zhang R, Jia D, Sun Y, Jia P. Circulating mediators linking cardiometabolic diseases to HFpEF: a mediation Mendelian randomization analysis. Cardiovasc Diabetol 2025; 24:201. [PMID: 40355922 PMCID: PMC12070650 DOI: 10.1186/s12933-025-02738-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Accepted: 04/10/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) is an increasingly prevalent clinical syndrome with high morbidity and mortality. Although HFpEF frequently coexists with cardiometabolic diseases, the causal mechanisms and potential mediators remain poorly understood. OBJECTIVES This study aimed to identify cardiometabolic risk factors specifically driving HFpEF and to determine their underlying circulating mediators. METHODS We used two-sample Mendelian Randomization (MR) to analyze the effects of obesity, Type 2 diabetes, hypertension, chronic kidney disease (CKD), and dyslipidemia on HFpEF and heart failure with reduced ejection fraction (HFrEF) in large European-ancestry GWAS datasets. We then performed mediation MR to identify plasma proteins and metabolites that mediate the transition from each cardiometabolic disease to HFpEF, respectively. We applied multivariable MR to assess the impact of risk confounding on the results. Bioinformatic analyses were conducted to delineate mechanisms. RESULTS Cardiometabolic diseases had heterogeneous effects on HFpEF and HFrEF. Obesity and type 2 diabetes showed adjusted causal effects with HFpEF, hypertension showed potential relevance to HFpEF, whereas dyslipidemia and CKD did not. MR analysis identified 5 proteins that mediate obesity to HFpEF; 5 proteins that mediate type 2 diabetes to HFpEF. Further mediation MR analysis of obesity and T2D on HFrEF revealed heterogeneity in circulating mediators between metabolic HFpEF and HFrEF. Comprehensive bioinformatics analyses showed that IL1R1, together with other proteins such as TP53 and FGF19, orchestrates the inflammatory and fibrotic processes underlying HFpEF. CONCLUSIONS These findings suggest that metabolic HFpEF has distinct etiological features compared with HFrEF and is driven by complex, condition-specific mediators. IL1R1 mediates HFpEF in multiple metabolic risk states, suggesting a potential therapeutic target. Further translational studies are warranted to evaluate anti-inflammatory strategies targeting IL1R1 in HFpEF.
Collapse
MESH Headings
- Humans
- Mendelian Randomization Analysis
- Heart Failure/genetics
- Heart Failure/physiopathology
- Heart Failure/blood
- Heart Failure/epidemiology
- Heart Failure/diagnosis
- Cardiometabolic Risk Factors
- Diabetes Mellitus, Type 2/genetics
- Diabetes Mellitus, Type 2/blood
- Diabetes Mellitus, Type 2/epidemiology
- Diabetes Mellitus, Type 2/diagnosis
- Risk Assessment
- Stroke Volume/genetics
- Biomarkers/blood
- Obesity/genetics
- Obesity/blood
- Obesity/epidemiology
- Obesity/diagnosis
- Dyslipidemias/genetics
- Dyslipidemias/blood
- Dyslipidemias/epidemiology
- Dyslipidemias/diagnosis
- Hypertension/genetics
- Hypertension/blood
- Hypertension/epidemiology
- Hypertension/diagnosis
- Mediation Analysis
- Ventricular Function, Left/genetics
- Renal Insufficiency, Chronic/genetics
- Renal Insufficiency, Chronic/blood
- Renal Insufficiency, Chronic/epidemiology
- Renal Insufficiency, Chronic/diagnosis
- Genetic Predisposition to Disease
- Phenotype
- Genome-Wide Association Study
- Databases, Genetic
Collapse
Affiliation(s)
- Mingzhi Lin
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Jiuqi Guo
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Hongqian Tao
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Zhilin Gu
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Wenyi Tang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Fuliang Zhou
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Yanling Jiang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Ruyi Zhang
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China
| | - Dalin Jia
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China.
| | - Yingxian Sun
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China.
- Key Laboratory of Environmental Stress and Chronic Disease Control and Prevention, Ministry of Education, China Medical University, Shenyang, Liaoning, China.
| | - Pengyu Jia
- Department of Cardiology, The First Hospital of China Medical University, 155 Nanjing North Street, Heping District, Shenyang, 110001, People's Republic of China.
| |
Collapse
|
6
|
Shen Y, Xie S, Lin Y, Fang Y, Zhang B, Zhang J. Maternal smoking around birth is associated with an increased risk of offspring constipation: Evidence from a Mendelian randomization study. Tob Induc Dis 2025; 23:TID-23-58. [PMID: 40342721 PMCID: PMC12060150 DOI: 10.18332/tid/203866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/07/2025] [Accepted: 04/10/2025] [Indexed: 05/11/2025] Open
Abstract
INTRODUCTION This study aimed to investigate the association between maternal smoking around birth and the incidence of offspring constipation. METHODS Genome-wide association study (GWAS) data for maternal smoking around birth and offspring constipation were obtained from the Mendelian randomization (MR) Base platform. Single nucleotide polymorphisms (SNPs) significantly associated with maternal smoking around birth were utilized as instrumental variables in two-sample MR analyses to explore the relationship between maternal smoking and offspring constipation. The analytical methods employed included the inverse-variance weighted (IVW) method, weighted median estimator, and MR-Egger regression. RESULTS Twenty SNPs significantly associated with maternal smoking around birth (p<5×10-8; linkage disequilibrium r2<0.001) were identified. Across the different methods, a consistent positive association was observed between maternal smoking around birth and an increased risk of constipation in offspring (IVW: OR=4.35; 95% CI: 1.81-10.45; weighted median estimator: OR=4.23; 95% CI: 1.22-14.75; MR-Egger: OR=0.92; 95% CI: 0.01-122.07), suggesting that higher frequency of maternal smoking is associated with an elevated risk of constipation in offspring. However, we did not detect any potential effect of genetic liability to constipation risk on maternal smoking. CONCLUSIONS This study provides evidence suggesting that increased maternal smoking around the time of birth may be linked to a higher risk of constipation in offspring.
Collapse
Affiliation(s)
- Yong Shen
- Fujian Children’s Hospital (Fujian Branch of Shanghai Children’s Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
- Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Siqi Xie
- Fujian Children’s Hospital (Fujian Branch of Shanghai Children’s Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Yu Lin
- Fujian Children’s Hospital (Fujian Branch of Shanghai Children’s Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Yifan Fang
- Fujian Children’s Hospital (Fujian Branch of Shanghai Children’s Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Bing Zhang
- Fujian Children’s Hospital (Fujian Branch of Shanghai Children’s Medical Center), College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| | - Jinna Zhang
- Fujian Maternity and Child Health Hospital College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, China
| |
Collapse
|
7
|
Chen S. Two-sample bi-directional causality between two traits with some invalid IVs in both directions using GWAS summary statistics. HGG ADVANCES 2025; 6:100449. [PMID: 40336198 DOI: 10.1016/j.xhgg.2025.100449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 05/01/2025] [Accepted: 05/01/2025] [Indexed: 05/09/2025] Open
Abstract
Mendelian randomization (MR) is a widely used method for assessing causal relationships between risk factors and outcomes using genetic variants as instrumental variables (IVs). While traditional MR assumes uni-directional causality, bi-directional MR aims to identify the true causal direction. In uni-directional MR, invalid IVs due to pleiotropy can violate assumptions and introduce biases. In bi-directional MR, traditional MR can be performed separately for each direction, but the presence of invalid IVs poses even greater challenges. We introduce a new bi-directional MR method incorporating stepwise selection (Bidir-SW) designed to address these challenges. Our approach leverages public genome-wide association study (GWAS) datasets for two traits and uses model selection criteria to identify invalid IVs iteratively by stepwise selection. This method accounts for potential bi-directional causality in the presence of common invalid IVs for both directions, even if only GWAS summary statistics are provided. Through simulation studies, we demonstrate that our method outperforms traditional MR techniques, such as MR-Egger and inverse-variance weighted (IVW), with uncorrelated SNPs. We also provide simulations to compare our approach with existing transcriptome-wide association study (TWAS) to show its effectiveness. Finally, we apply the proposed method to genetic traits such as CRP levels and BMI to explore possible bi-directional relationships among these traits. We also used the proposed method to discover causal protein biomarkers. Our findings suggest that the Bidir-SW approach is a powerful tool for bi-directional MR or TWAS, which can provide a valuable framework for future genetic epidemiology studies.
Collapse
Affiliation(s)
- Siyi Chen
- School of Public Health, LSU Health Sciences Center New Orleans, New Orleans, LA 70112, USA.
| |
Collapse
|
8
|
Ma F, Longo M, Meroni M, Bhattacharya D, Paolini E, Mughal S, Hussain S, Anand SK, Gupta N, Zhu Y, Navarro-Corcuera A, Li K, Prakash S, Cogliati B, Wang S, Huang X, Wang X, Yurdagul A, Rom O, Wang L, Fried SK, Dongiovanni P, Friedman SL, Cai B. EHBP1 suppresses liver fibrosis in metabolic dysfunction-associated steatohepatitis. Cell Metab 2025; 37:1152-1170.e7. [PMID: 40015280 PMCID: PMC12058419 DOI: 10.1016/j.cmet.2025.01.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 11/27/2024] [Accepted: 01/22/2025] [Indexed: 03/01/2025]
Abstract
Excess cholesterol accumulation contributes to fibrogenesis in metabolic dysfunction-associated steatohepatitis (MASH), but how hepatic cholesterol metabolism becomes dysregulated in MASH is not completely understood. We show that human fibrotic MASH livers have decreased EH-domain-binding protein 1 (EHBP1), a genome-wide association study (GWAS) locus associated with low-density lipoprotein (LDL) cholesterol, and that EHBP1 loss- and gain-of-function increase and decrease MASH fibrosis in mice, respectively. Mechanistic studies reveal that EHBP1 promotes sortilin-mediated PCSK9 secretion, leading to LDL receptor (LDLR) degradation, decreased LDL uptake, and reduced TAZ, a fibrogenic effector. At a cellular level, EHBP1 deficiency affects the intracellular localization of retromer, a protein complex required for sortilin stabilization. Our therapeutic approach to stabilizing retromer is effective in mitigating MASH fibrosis. Moreover, we show that the tumor necrosis factor alpha (TNF-α)/peroxisome proliferator-activated receptor alpha (PPARα) pathway suppresses EHBP1 in MASH. These data not only provide mechanistic insights into the role of EHBP1 in cholesterol metabolism and MASH fibrosis but also elucidate an interplay between inflammation and EHBP1-mediated cholesterol metabolism.
Collapse
Affiliation(s)
- Fanglin Ma
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Miriam Longo
- Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Marica Meroni
- Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Dipankar Bhattacharya
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Erika Paolini
- Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Shama Mughal
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Syed Hussain
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sumit Kumar Anand
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA 71103, USA
| | - Neha Gupta
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yiwei Zhu
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Amaia Navarro-Corcuera
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kenneth Li
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Satya Prakash
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bruno Cogliati
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Shuang Wang
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xin Huang
- Columbia Center for Human Development, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Xiaobo Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA 71103, USA
| | - Oren Rom
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA 71103, USA
| | - Liheng Wang
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Susan K Fried
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Paola Dongiovanni
- Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Scott L Friedman
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bishuang Cai
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
9
|
Cheng X, Li L, Lin X, Chen N, Liu X, Li Y, Li Z, Gong J, Liu Q, Wang Y, Wang J, Xia Z, Lu Y, Jin H, Zhang X, Wang L, Chen J, Fan G, Deng S, Zhao S, Zhu L. Developing a polygenic risk score for pelvic organ prolapse: a combined risk assessment approach in Chinese women. Front Med 2025:10.1007/s11684-024-1114-2. [PMID: 40317452 DOI: 10.1007/s11684-024-1114-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 09/18/2024] [Indexed: 05/07/2025]
Abstract
Pelvic organ prolapse (POP), whose etiology is influenced by genetic and clinical risk factors, considerably impacts women's quality of life. However, the genetic underpinnings in non-European populations and comprehensive risk models integrating genetic and clinical factors remain underexplored. This study constructed the first polygenic risk score (PRS) for POP in the Chinese population by utilizing 20 disease-associated variants from the largest existing genome-wide association study. We analyzed a discovery cohort of 576 cases and 623 controls and a validation cohort of 264 cases and 200 controls. Results showed that the case group exhibited a significantly higher PRS than the control group. Moreover, the odds ratio of the top 10% risk group was 2.6 times higher than that of the bottom 10%. A high PRS was significantly correlated with POP occurrence in women older than 50 years old and in those with one or no childbirths. As far as we know, the integrated prediction model, which combined PRS and clinical risk factors, demonstrated better predictive accuracy than other existing PRS models. This combined risk assessment model serves as a robust tool for POP risk prediction and stratification, thereby offering insights into individualized preventive measures and treatment strategies in future clinical practice.
Collapse
Affiliation(s)
- Xi Cheng
- Department of Obstetrics and Gynecology National Clinical Research Center for Obstetric & Gynecologic Diseases, State Key Laboratory of Common Mechanism Research for Major Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Lei Li
- Department of Obstetrics and Gynecology National Clinical Research Center for Obstetric & Gynecologic Diseases, State Key Laboratory of Common Mechanism Research for Major Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Xijuan Lin
- Department of Obstetrics and Gynecology National Clinical Research Center for Obstetric & Gynecologic Diseases, State Key Laboratory of Common Mechanism Research for Major Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Na Chen
- Department of Obstetrics and Gynecology National Clinical Research Center for Obstetric & Gynecologic Diseases, State Key Laboratory of Common Mechanism Research for Major Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Xudong Liu
- The State Key Laboratory for Complex, Severe, and Rare Diseases, Peking Union Medical College Hospital, Beijing, 100730, China
| | - Yaqian Li
- Clinical Research Institute, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Zhaoai Li
- Department of Gynecology and Obstetrics, Children's Hospital of Shanxi Province, Taiyuan, 030013, China
| | - Jian Gong
- Department of Gynecology and Obstetrics, Maternal and Child Health Hospital of Wuxi, Wuxi, 214002, China
| | - Qing Liu
- Department of Gynecology and Obstetrics, Maternal and Child Health Hospital of Gansu Province, Lanzhou, 730030, China
| | - Yuling Wang
- Department of Gynecology and Obstetrics, Maternal and Child Health Hospital of Foshan, Foshan, 528000, China
| | - Juntao Wang
- Department of Gynecology and Obstetrics, Maternal and Child Health Hospital of Guiyang, Guiyang, 550003, China
| | - Zhijun Xia
- Department of Gynecology and Obstetrics, Sheng Jing Hospital of China Medical University, Shenyang, 117004, China
| | - Yongxian Lu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chinese PLA General Hospital, Beijing, 100037, China
| | - Hangmei Jin
- Department of Obstetrics and Gynecology, Women's Hospital, Zhejiang University, Hangzhou, 310006, China
| | - Xiaowei Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Luwen Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Juan Chen
- Department of Obstetrics and Gynecology National Clinical Research Center for Obstetric & Gynecologic Diseases, State Key Laboratory of Common Mechanism Research for Major Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Guorong Fan
- Department of Obstetrics and Gynecology National Clinical Research Center for Obstetric & Gynecologic Diseases, State Key Laboratory of Common Mechanism Research for Major Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Shan Deng
- Department of Obstetrics and Gynecology National Clinical Research Center for Obstetric & Gynecologic Diseases, State Key Laboratory of Common Mechanism Research for Major Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Sen Zhao
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, 77030, USA.
| | - Lan Zhu
- Department of Obstetrics and Gynecology National Clinical Research Center for Obstetric & Gynecologic Diseases, State Key Laboratory of Common Mechanism Research for Major Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China.
| |
Collapse
|
10
|
Giuranna J, Zheng Y, Brandt M, Jall S, Mukherjee A, Shankhwar S, Renner S, Kurapati NK, May C, Peters T, Herpertz-Dahlmann B, Seitz J, de Zwaan M, Herzog W, Ehrlich S, Zipfel S, Giel K, Egberts K, Burghardt R, Föcker M, Marcus K, Keyvani K, Müller TD, Schmitz F, Rajcsanyi LS, Hinney A. Genetic and functional analyses of CTBP2 in anorexia nervosa and body weight regulation. Mol Psychiatry 2025; 30:1836-1846. [PMID: 39511451 PMCID: PMC12014503 DOI: 10.1038/s41380-024-02791-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 11/15/2024]
Abstract
The C-terminal binding protein 2 (CTBP2) gene (translational isoforms: CTBP2-L/S, RIBEYE) had been identified by a cross-trait analysis of genome-wide association studies for anorexia nervosa (AN) and body mass index (BMI). Here, we did a mutation analysis in CTBP2 by performing polymerase chain reactions with subsequent Sanger-sequencing to identify variants relevant for AN and body weight regulation and ensued functional studies. Analysis of the coding regions of CTBP2 in 462 female patients with AN (acute or recovered), 490 children and adolescents with severe obesity, 445 healthy-lean adult individuals and 168 healthy adult individuals with normal body weight detected 24 variants located in the specific exon of RIBEYE. In the initial analysis, three of these were rare non-synonymous variants (NSVs) detected heterozygously in patients with AN (p.Arg72Trp - rs146900874; p.Val289Met -rs375685611 and p.Gly362Arg - rs202010294). Four NSVs and one heterozygous frameshift variant were exclusively detected in children and adolescents with severe obesity (p.Pro53Ser - rs150867595; p.Gln175ArgfsTer45 - rs141864737; p.Leu310Val - rs769811964; p.Pro397Ala - rs76134089 and p.Pro402Ser - rs113477585). Ribeye mRNA was detected in mouse hypothalamus. No effect of fasting or overfeeding on murine hypothalamic Ribeye expression was determined. Yet, increased Ribeye expression was detected in hypothalami of leptin-treated Lepob/ob mice. This increase was not related to reduced food intake and leptin-induced weight loss. We detected rare and frequent variants in the RIBEYE specific exon in both patients with AN and in children and adolescents with severe obesity. Our data suggest RIBEYE as a relevant gene for weight regulation.
Collapse
Affiliation(s)
- Johanna Giuranna
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, Germany
| | - Yiran Zheng
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, Germany
- Helmholtz Pioneer Campus, Helmholtz Zentrum München, Munich, Germany
| | | | - Sigrid Jall
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Amrita Mukherjee
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Medical School, Saarland University, Homburg, Germany
| | - Soni Shankhwar
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Medical School, Saarland University, Homburg, Germany
| | - Simone Renner
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Institute of Molecular Animal Breeding and Biotechnology, Ludwig-Maximilian University Munich (LMU), Munich, Germany
| | - Nirup Kumar Kurapati
- Institute of Neuropathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Caroline May
- Medizinisches Proteom-Center, Ruhr-University Bochum, Bochum, Germany
| | - Triinu Peters
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, Germany
- Section of Molecular Genetics in Mental Disorders, University Hospital Essen, Essen, Germany
- Institute of Sex and Gender-Sensitive Medicine, University Hospital Essen, Essen, Germany
| | - Beate Herpertz-Dahlmann
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Jochen Seitz
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Department of Child and Adolescent Psychiatry and Psychotherapy, University Hospital of the RWTH Aachen, Aachen, Germany
| | - Martina de Zwaan
- Department of Psychosomatic Medicine and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Wolfgang Herzog
- Department of Internal Medicine II, General Internal and Psychosomatic Medicine, University of Heidelberg, Heidelberg, Germany
| | - Stefan Ehrlich
- Eating Disorders Research and Treatment Center, Department of Child and Adolescent Psychiatry, Faculty of Medicine, TU Dresden, Dresden, Germany
- Translational Developmental Neuroscience Section, Division of Psychological and Social Medicine and Developmental Neurosciences, Faculty of Medicine, TU Dresden, Germany
| | - Stephan Zipfel
- Department of Psychosomatic Medicine and Psychotherapy, Medical University Hospital Tübingen, Tübingen, Germany
- Center of Excellence in Eating Disorders KOMET, Tübingen, Germany
- German Center for Mental Health (DZPG), Tübingen, Germany
| | - Katrin Giel
- Department of Psychosomatic Medicine and Psychotherapy, Medical University Hospital Tübingen, Tübingen, Germany
- Center of Excellence in Eating Disorders KOMET, Tübingen, Germany
- German Center for Mental Health (DZPG), Tübingen, Germany
| | - Karin Egberts
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University of Würzburg, Würzburg, Germany
| | - Roland Burghardt
- Child and Adolescent Psychiatry Clinic, Oberberg Fachklinik Fasanenkiez Berlin, Berlin, Germany
| | - Manuel Föcker
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Münster, Münster, Germany
- LWL-University Hospital Hamm for Child and Adolescent Psychiatry, Ruhr-University Bochum, Hamm, Germany
| | - Katrin Marcus
- Medizinisches Proteom-Center, Ruhr-University Bochum, Bochum, Germany
| | - Kathy Keyvani
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, Germany
- Institute of Neuropathology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Walther-Straub-Insitute for Pharmacology and Toxicology, Ludwig-Maximilians University Munich (LMU), Munich, Germany
| | - Frank Schmitz
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Medical School, Saarland University, Homburg, Germany
| | - Luisa Sophie Rajcsanyi
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, Germany.
- Section of Molecular Genetics in Mental Disorders, University Hospital Essen, Essen, Germany.
- Institute of Sex and Gender-Sensitive Medicine, University Hospital Essen, Essen, Germany.
| | - Anke Hinney
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, Essen, Germany
- Section of Molecular Genetics in Mental Disorders, University Hospital Essen, Essen, Germany
- Institute of Sex and Gender-Sensitive Medicine, University Hospital Essen, Essen, Germany
| |
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
|
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
|
13
|
Lin J, Lei L, Liang Q, Huang X, Ding Y, Pan L, Yang J, Li W. Assessment of causality association between serum adiponectin levels and the risk of Alzheimer's disease and Parkinson's disease: a Mendelian randomization study. Front Neurol 2025; 16:1395798. [PMID: 40371086 PMCID: PMC12075267 DOI: 10.3389/fneur.2025.1395798] [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: 03/05/2024] [Accepted: 04/11/2025] [Indexed: 05/16/2025] Open
Abstract
Background Until recently, the association between circulating adiponectin (ADPN) levels and the risk of Alzheimer's disease (AD) and Parkinson's disease (PD) remained unclear. Methods We utilized public data from the IEU GWAS database to conduct a two-sample bidirectional Mendelian randomization (MR) analysis and multiple sensitivity analyses. The MR analysis was performed using the aggregated data, with the genetic risk score (GRS) serving as an instrumental variable. Results The MR analyses revealed no significant causal association between genetically determined ADPN levels and the risk of AD (ORIVW = 0.852, 95% confidence interval [CI]: 0.586-1.117, p = 0.235) or PD (ORIVW = 0.830, 95% CI: 0.780-1.156, p = 0.606). Conversely, neither AD nor PD demonstrated any causal association with ADPN levels. The GRS approach yielded similar results (p > 0.05). However, it exhibited a negative correlation with interleukin 1β (IL1β, βIVW = -0.31; 95% CI: -0.55 to -0.07, p = 0.011). The Cochrane's Q test and MR-PRESSO analysis revealed no evidence of pleiotropy. Conclusion Our findings provide no evidence to substantiate a causal relationship between ADPN levels and the risk of AD and PD or vice versa. However, elevated levels of ADPN may correlate with lower levels of IL1β.
Collapse
Affiliation(s)
- Jiali Lin
- Research Center of Health Management, Guangxi Zhuang Autonomous Region People's Hospital, Guangxi Academy of Medical Sciences, Nanning, China
| | - Langhuan Lei
- Research Center of Health Management, Guangxi Zhuang Autonomous Region People's Hospital, Guangxi Academy of Medical Sciences, Nanning, China
| | - Qiuyu Liang
- Research Center of Health Management, Guangxi Zhuang Autonomous Region People's Hospital, Guangxi Academy of Medical Sciences, Nanning, China
| | - Xiaozhi Huang
- Department of Health Management, Guangxi Zhuang Autonomous Region People's Hospital, Guangxi Academy of Medical Sciences, Nanning, China
| | - Yanping Ding
- Department of Health Management, Guangxi Zhuang Autonomous Region People's Hospital, Guangxi Academy of Medical Sciences, Nanning, China
| | - Liuxian Pan
- Department of Health Management, Guangxi Zhuang Autonomous Region People's Hospital, Guangxi Academy of Medical Sciences, Nanning, China
| | - Jianrong Yang
- Research Center of Health Management, Guangxi Zhuang Autonomous Region People's Hospital, Guangxi Academy of Medical Sciences, Nanning, China
| | - Wei Li
- Research Center of Health Management, Guangxi Zhuang Autonomous Region People's Hospital, Guangxi Academy of Medical Sciences, Nanning, China
- Department of Health Management, Guangxi Zhuang Autonomous Region People's Hospital, Guangxi Academy of Medical Sciences, Nanning, China
| |
Collapse
|
14
|
Jia G, Guo T, Liu L, He C. Causal relationship between serum urate and asthma: a Mendelian randomization study. J Asthma 2025:1-9. [PMID: 40262517 DOI: 10.1080/02770903.2025.2495734] [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: 02/06/2025] [Revised: 04/08/2025] [Accepted: 04/16/2025] [Indexed: 04/24/2025]
Abstract
BACKGROUND Previous studies have suggested that elevated urate levels may increase the risk of asthma; however, the nature of this association remains debated. To clarify this uncertainty, we conducted a Mendelian randomization (MR) study to investigate the potential causal relationship. METHODS Summary statistics for urate were sourced from the Global Urate Genetics Consortium (GUGC), and those for asthma were obtained from the FinnGen database. Genetic variants strongly associated with urate were selected as instrumental variables (IVs). Univariable and multivariable MR analyses were conducted to investigate the causal relationship between urate and asthma. Subsequently, network MR analyses were performed to reveal the mediating role of urate in the relationship between body mass index (BMI) and asthma. RESULTS The univariable MR analysis showed that urate was associated with an increased risk of asthma (IVW OR = 1.13, 95%CI = 1.04-1.23, p = 0.004). This causal relationship remained consistent in multivariable MR analyses, even after adjusting for potential confounders, including smoking initiation, cigarettes per day, alcohol intake frequency, BMI, allergic rhinitis, and gastroesophageal reflux disease (GERD). Furthermore, network MR analyses demonstrated that the proportion of causal effect between BMI and asthma mediated by urate was 18.05% (95%CI = 6.23%-29.88%). CONCLUSION Our study confirms that serum urate is associated with an increased risk of asthma, suggesting its potential as a target for both prevention and treatment. Additionally, our findings indicate that urate partially mediates the relationship between BMI and asthma, emphasizing its role in the mechanism underlying BMI-induced asthma.
Collapse
Affiliation(s)
- Guobing Jia
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Respiratory and Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tao Guo
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lei Liu
- Department of Respiratory and Critical Care Medicine, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Chengshi He
- Department of Respiratory and Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
15
|
Goodwill JR, Taylor HO. Measuring Whether Gratitude and Loneliness Mediate the Link Between Non-organizational Religiosity and Suicidal Ideation: Evidence From Black Adults During COVID-19. Public Health Rep 2025:333549251314665. [PMID: 40296509 PMCID: PMC12040851 DOI: 10.1177/00333549251314665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2025] Open
Abstract
OBJECTIVES Suicides among Black adults in the United States increased nationally during the COVID-19 pandemic, although limited empirical evidence documents the pathways that explain how suicide risk may develop in this population. We examined experiences of non-organizational religious involvement, gratitude, and loneliness and their relation to suicidal ideation among Black adults in the United States. METHODS We analyzed data from a probability-based sample of 995 Black adults in the United States who completed online surveys from April through June 2022. We recruited participants from the AmeriSpeak panel at the National Opinion Research Center. We applied structural equation modeling techniques to measure direct and indirect associations among religiosity, positive psychology, and mental health variables. We tested whether non-organizational religiosity was indirectly associated with suicidal ideation via feelings of gratitude and COVID-19-specific forms of loneliness during the pandemic. RESULTS The measurement model demonstrated a good fit to the data. Structural model results indicated that non-organizational religious involvement was positively related to gratitude (β = 0.51; P < .001); in turn, feelings of gratitude were associated with reduced suicidal ideation (β = -0.12; P = .02). Moreover, COVID-19-specific forms of loneliness were positively associated with past-year suicidal ideation (β = 0.11; P = .01). Non-organizational religious involvement, however, was not directly associated with feelings of COVID-19-related loneliness or suicidal ideation. CONCLUSIONS Public health officials should account for feelings of gratitude and loneliness as mechanisms that can be leveraged to inform the development of evidence-based suicide prevention interventions for Black adults during public health emergencies such as the COVID-19 pandemic and beyond.
Collapse
Affiliation(s)
- Janelle R. Goodwill
- Crown Family School of Social Work, Policy, and Practice, University of Chicago, Chicago, IL, USA
| | - Harry O. Taylor
- Factor-Inwentash Faculty of Social Work, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
16
|
Verde L, Galasso M, Coletta DK, Savastano S, Mandarino LJ, Colao A, Barrea L, Muscogiuri G. The Interplay of UCP3 and PCSK1 Variants in Severe Obesity. Curr Obes Rep 2025; 14:38. [PMID: 40281302 PMCID: PMC12031958 DOI: 10.1007/s13679-025-00631-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/16/2025] [Indexed: 04/29/2025]
Abstract
Obesity is a heterogeneous and multifactorial disease with a strong genetic component. While polygenic obesity accounts for most common cases, rare monogenic variants contribute, particularly in severe, early-onset obesity. Among the lesser-studied candidates are UCP3 and PCSK1, genes involved in key metabolic pathways. RECENT FINDINGS: The UCP3 p.Val192Ile (c.574G > A) and PCSK1 p.Asn221Asp (c.661 A > G) variants have been independently associated with metabolic pathways, including fatty acid oxidation and hormone processing, as well as a modestly increased risk of obesity. Clinical and genetic characterization of two patients with severe early-onset obesity revealed the co-occurrence of these variants, which were associated with metabolic disturbances such as insulin resistance. PURPOSE OF THE REVIEW: This narrative review examined the functional and clinical significance of UCP3 and PCSK1 variants in severe obesity, presenting two case reports to illustrate their potential impact. Our findings support a potential model in which rare variants in distinct metabolic genes may interact synergistically to exacerbate disease severity. Further studies are needed to elucidate their combined functional effects and contributions to obesity pathogenesis.
Collapse
Affiliation(s)
- Ludovica Verde
- Department of Public Health, University of Naples Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ, USA
| | - Martina Galasso
- Dipartimento di Medicina Clinica e Chirurgia, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Dawn K Coletta
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ, USA
- Department of Physiology, University of Arizona, Tucson, AZ, USA
- Center for Disparities in Diabetes, Obesity and Metabolism, University of Arizona, Tucson, AZ, USA
| | - Silvia Savastano
- Dipartimento di Medicina Clinica e Chirurgia, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Unità di Endocrinologia, Diabetologia e Andrologia, Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
| | - Lawrence J Mandarino
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ, USA
- Center for Disparities in Diabetes, Obesity and Metabolism, University of Arizona, Tucson, AZ, USA
| | - Annamaria Colao
- Dipartimento di Medicina Clinica e Chirurgia, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Unità di Endocrinologia, Diabetologia e Andrologia, Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy
- Cattedra Unesco "Educazione Alla Salute E Allo Sviluppo Sostenibile", University Federico II, 80131, Naples, Italy
| | - Luigi Barrea
- Dipartimento di Psicologia e Scienze della Salute, , Università Telematica Pegaso, Centro Direzionale Isola F2, Via Porzio, Isola F2, 80143, Naples, Italy
| | - Giovanna Muscogiuri
- Department of Medicine, Division of Endocrinology, University of Arizona, Tucson, AZ, USA.
- Dipartimento di Medicina Clinica e Chirurgia, Centro Italiano per la cura e il Benessere del Paziente con Obesità (C.I.B.O), Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy.
- Unità di Endocrinologia, Diabetologia e Andrologia, Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli Federico II, Via Sergio Pansini 5, 80131, Naples, Italy.
- Cattedra Unesco "Educazione Alla Salute E Allo Sviluppo Sostenibile", University Federico II, 80131, Naples, Italy.
| |
Collapse
|
17
|
Xu L, Zhou G, Jiang W, Zhang H, Dong Y, Guan L, Zhao H. JointPRS: A data-adaptive framework for multi-population genetic risk prediction incorporating genetic correlation. Nat Commun 2025; 16:3841. [PMID: 40268942 PMCID: PMC12019179 DOI: 10.1038/s41467-025-59243-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 04/16/2025] [Indexed: 04/25/2025] Open
Abstract
Genetic risk prediction for non-European populations is hindered by limited Genome-Wide Association Study (GWAS) sample sizes and small tuning datasets. We propose JointPRS, a data-adaptive framework that leverages genetic correlations across multiple populations using GWAS summary statistics. It achieves accurate predictions without individual-level tuning data and remains effective in the presence of a small tuning set thanks to its data-adaptive approach. Through extensive simulations and real data applications to 22 quantitative and four binary traits in five continental populations evaluated using the UK Biobank (UKBB) and All of Us (AoU), JointPRS consistently outperforms six state-of-the-art methods across three data scenarios: no tuning data, same-cohort tuning and testing, and cross-cohort tuning and testing. Notably, in the Admixed American population, JointPRS improves lipid trait prediction in AoU by 6.46%-172.00% compared to the other existing methods.
Collapse
Affiliation(s)
- Leqi Xu
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Geyu Zhou
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Wei Jiang
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
- Department of Mathematics, University of Texas at Arlington, Arlington, Texas, USA
- Division of Data Science, College of Science, University of Texas at Arlington, Arlington, Texas, USA
| | - Haoyu Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Yikai Dong
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA
| | - Leying Guan
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA.
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA.
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, New Haven, CT, USA.
- Program in Computational Biology and Bioinformatics, Yale University, New Haven, CT, USA.
| |
Collapse
|
18
|
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
|
19
|
Zhou G, Qie X, Zhao H. A Bayesian approach to correcting the attenuation bias of regression using polygenic risk score. Genetics 2025; 229:iyaf018. [PMID: 39891671 DOI: 10.1093/genetics/iyaf018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/08/2025] [Accepted: 01/21/2025] [Indexed: 02/03/2025] Open
Abstract
Polygenic risk score has become increasingly popular for predicting the value of complex traits. In many settings, polygenic risk score is used as a covariate in regression analysis to study the association between different phenotypes. However, measurement error in polygenic risk score causes attenuation bias in the estimation of regression coefficients. In this paper, we employ a Bayesian approach to accounting for the measurement error of polygenic risk score and correcting the attenuation bias in linear and logistic regression. Through simulation, we show that our approach is able to obtain approximately unbiased estimation of coefficients and credible intervals with correct coverage probability. We also empirically compare our Bayesian measurement error model with the conventional regression model by analyzing real traits in the UK Biobank. The results demonstrate the effectiveness of our approach as it significantly reduces the error in coefficient estimates.
Collapse
Affiliation(s)
- Geyu Zhou
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT 06511, USA
| | - Xinyue Qie
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT 06511, USA
| | - Hongyu Zhao
- Department of Biostatistics, Yale School of Public Health, Yale University, New Haven, CT 06511, USA
| |
Collapse
|
20
|
Clifford D, Laing E, Harris C, Slagel N, Levinson J, Squires N, Hunger J. Incorporating Weight-Inclusive Approaches in Higher Education Curriculum for Future Nutrition and Dietetics Professionals. JOURNAL OF NUTRITION EDUCATION AND BEHAVIOR 2025:S1499-4046(25)00092-2. [PMID: 40243955 DOI: 10.1016/j.jneb.2025.03.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 03/14/2025] [Accepted: 03/23/2025] [Indexed: 04/18/2025]
Abstract
While weight-inclusive approaches are becoming more widely accepted and used in nutrition and dietetics, educators and professionals often lack adequate training. The purpose of this paper is to provide faculty and preceptors with strategies to incorporate weight-inclusive approaches into nutrition and dietetics curricula. We present a framework for demonstrating how students can meet knowledge and competency requirements embedded in courses in nationally accredited nutrition and dietetics programs using a variety of weight-inclusive concepts and learning activities. Providing weight-inclusive approaches is essential for equipping nutrition professionals to offer patient-centered care that minimizes weight stigma and disordered eating.
Collapse
Affiliation(s)
- Dawn Clifford
- Department of Health Sciences, Northern Arizona University, Flagstaff, AZ.
| | - Emma Laing
- Department of Nutritional Sciences, University of Georgia, Athens, GA
| | - Cristen Harris
- Department of Epidemiology, University of Washington, Seattle, WA
| | - Nicholas Slagel
- Department of Kinesiology, Nutrition, and Dietetics, University of Northern Colorado, Greeley, CO
| | - Jordan Levinson
- Department of Nutrition and Food Sciences, University of Vermont, Burlington, VT
| | - Nikole Squires
- Department of Health Sciences, Northern Arizona University, Flagstaff, AZ
| | | |
Collapse
|
21
|
Wang Y, Chen Y, Liu Y, Chen S. Molecular Mechanism of the Grid Gene Family Regulating Growth Size Heteromorphism in Cynoglossus semilaevis. Animals (Basel) 2025; 15:1130. [PMID: 40281964 PMCID: PMC12024286 DOI: 10.3390/ani15081130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/29/2025] Open
Abstract
There are obvious individual differences in the growth and development of Cynoglossus semilaevis, mainly due to female bias. We selected 500 female Cynoglossus semilaevis of different sizes for GWAS and transcriptome analysis to screen for differential genes. qPCR was performed to detect the expression of the genes in various tissues, and RNAi experiments were performed in testicular cells to knock down the grid1 and grid2 genes and transcriptome sequencing was performed to check the changes of the downstream genes. Grid gene was screened for the common genes by GWAS sequencing and transcriptome sequencing. In the QPCR results, the expression of the grid gene family was negatively correlated with fish size, and was slightly higher in males than in females; in the transcriptome results, the expression of shcbp1, sass6, cdca7, and gh was up-regulated, and the expression of igf1 was down-regulated. It is speculated that igf1 has an antagonistic effect on gh, which is deregulated when the grid gene family is knocked down. The grid gene family may affect the growth of individual Cynoglossus semilaevis through the gh-igf1 axis, which provides a basis for the study of the differences in the growth size of Cynoglossus semilaevis.
Collapse
Affiliation(s)
- Yaning Wang
- College of Life Science, Qingdao University, Qingdao 266071, China;
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; (Y.C.); (Y.L.)
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Yadong Chen
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; (Y.C.); (Y.L.)
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Yang Liu
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; (Y.C.); (Y.L.)
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| | - Songlin Chen
- State Key Laboratory of Mariculture Biobreeding and Sustainable Goods, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao 266071, China; (Y.C.); (Y.L.)
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao 266237, China
| |
Collapse
|
22
|
Zhang X, Brody JA, Graff M, Highland HM, Chami N, Xu H, Wang Z, Ferrier KR, Chittoor G, Josyula NS, Meyer M, Gupta S, Li X, Li Z, Allison MA, Becker DM, Bielak LF, Bis JC, Boorgula MP, Bowden DW, Broome JG, Buth EJ, Carlson CS, Chang KM, Chavan S, Chiu YF, Chuang LM, Conomos MP, DeMeo DL, Du M, Duggirala R, Eng C, Fohner AE, Freedman BI, Garrett ME, Guo X, Haiman C, Heavner BD, Hidalgo B, Hixson JE, Ho YL, Hobbs BD, Hu D, Hui Q, Hwu CM, Jackson RD, Jain D, Kalyani RR, Kardia SLR, Kelly TN, Lange EM, LeNoir M, Li C, Le Marchand L, McDonald MLN, McHugh CP, Morrison AC, Naseri T, O'Connell J, O'Donnell CJ, Palmer ND, Pankow JS, Perry JA, Peters U, Preuss MH, Rao DC, Regan EA, Reupena SM, Roden DM, Rodriguez-Santana J, Sitlani CM, Smith JA, Tiwari HK, Vasan RS, Wang Z, Weeks DE, Wessel J, Wiggins KL, Wilkens LR, Wilson PWF, Yanek LR, Yoneda ZT, Zhao W, Zöllner S, Arnett DK, Ashley-Koch AE, Barnes KC, Blangero J, Boerwinkle E, Burchard EG, Carson AP, Chasman DI, Ida Chen YD, Curran JE, Fornage M, Gordeuk VR, He J, Heckbert SR, Hou L, Irvin MR, et alZhang X, Brody JA, Graff M, Highland HM, Chami N, Xu H, Wang Z, Ferrier KR, Chittoor G, Josyula NS, Meyer M, Gupta S, Li X, Li Z, Allison MA, Becker DM, Bielak LF, Bis JC, Boorgula MP, Bowden DW, Broome JG, Buth EJ, Carlson CS, Chang KM, Chavan S, Chiu YF, Chuang LM, Conomos MP, DeMeo DL, Du M, Duggirala R, Eng C, Fohner AE, Freedman BI, Garrett ME, Guo X, Haiman C, Heavner BD, Hidalgo B, Hixson JE, Ho YL, Hobbs BD, Hu D, Hui Q, Hwu CM, Jackson RD, Jain D, Kalyani RR, Kardia SLR, Kelly TN, Lange EM, LeNoir M, Li C, Le Marchand L, McDonald MLN, McHugh CP, Morrison AC, Naseri T, O'Connell J, O'Donnell CJ, Palmer ND, Pankow JS, Perry JA, Peters U, Preuss MH, Rao DC, Regan EA, Reupena SM, Roden DM, Rodriguez-Santana J, Sitlani CM, Smith JA, Tiwari HK, Vasan RS, Wang Z, Weeks DE, Wessel J, Wiggins KL, Wilkens LR, Wilson PWF, Yanek LR, Yoneda ZT, Zhao W, Zöllner S, Arnett DK, Ashley-Koch AE, Barnes KC, Blangero J, Boerwinkle E, Burchard EG, Carson AP, Chasman DI, Ida Chen YD, Curran JE, Fornage M, Gordeuk VR, He J, Heckbert SR, Hou L, Irvin MR, Kooperberg C, Minster RL, Mitchell BD, Nouraie M, Psaty BM, Raffield LM, Reiner AP, Rich SS, Rotter JI, Benjamin Shoemaker M, Smith NL, Taylor KD, Telen MJ, Weiss ST, Zhang Y, Heard-Costa N, Sun YV, Lin X, Cupples LA, Lange LA, Liu CT, Loos RJF, North KE, Justice AE. Whole genome sequencing analysis of body mass index identifies novel African ancestry-specific risk allele. Nat Commun 2025; 16:3470. [PMID: 40216759 PMCID: PMC11992084 DOI: 10.1038/s41467-025-58420-2] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Obesity is a major public health crisis associated with high mortality rates. Previous genome-wide association studies (GWAS) investigating body mass index (BMI) have largely relied on imputed data from European individuals. This study leveraged whole-genome sequencing (WGS) data from 88,873 participants from the Trans-Omics for Precision Medicine (TOPMed) Program, of which 51% were of non-European population groups. We discovered 18 BMI-associated signals (P < 5 × 10-9), including two secondary signals. Notably, we identified and replicated a novel low-frequency single nucleotide polymorphism (SNP) in MTMR3 that was common in individuals of African descent. Using a diverse study population, we further identified two novel secondary signals in known BMI loci and pinpointed two likely causal variants in the POC5 and DMD loci. Our work demonstrates the benefits of combining WGS and diverse cohorts in expanding current catalog of variants and genes confer risk for obesity, bringing us one step closer to personalized medicine.
Collapse
Affiliation(s)
- Xinruo Zhang
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Mariaelisa Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Heather M Highland
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Nathalie Chami
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hanfei Xu
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Zhe Wang
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kendra R Ferrier
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | | | | | - Mariah Meyer
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Shreyash Gupta
- Population Health Sciences, Geisinger, Danville, PA, USA
| | - Xihao Li
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Genetics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Zilin Li
- Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN, USA
- School of Mathematics and Statistics and KLAS, Northeast Normal University, Changchun, Jilin, China
| | - Matthew A Allison
- Department of Family Medicine, Division of Preventive Medicine, The University of California San Diego, La Jolla, CA, USA
| | - Diane M Becker
- Department of Medicine, General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lawrence F Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Joshua C Bis
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | | | - Donald W Bowden
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jai G Broome
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
- Department of Medicine, Division of Medical Genetics, University of Washington, Seattle, WA, USA
| | - Erin J Buth
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | | | - Kyong-Mi Chang
- The Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA, USA
- University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sameer Chavan
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Yen-Feng Chiu
- Institute of Population Health Sciences, National Health Research Institutes, Taipei, Taiwan
| | - Lee-Ming Chuang
- Department of Internal Medicine, Division of Metabolism/Endocrinology, National Taiwan University Hospital, Taipei, Taiwan
| | - Matthew P Conomos
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Dawn L DeMeo
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Mengmeng Du
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ravindranath Duggirala
- Life Sciences, College of Arts and Sciences, Texas A&M University-San Antonio, San Antonio, TX, USA
- Department of Health and Behavioral Sciences, College of Arts and Sciences, Texas A&M University-San Antonio, San Antonio, TX, USA
| | - Celeste Eng
- Department of Medicine, Lung Biology Center, University of California, San Francisco, San Francisco, CA, USA
| | - Alison E Fohner
- Epidemiology, Institute of Public Health Genetics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Barry I Freedman
- Internal Medicine, Section on Nephrology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Melanie E Garrett
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Xiuqing Guo
- Department of Pediatrics, Genomic Outcomes, The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Chris Haiman
- Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Benjamin D Heavner
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Bertha Hidalgo
- Department of Epidemiology, School of Public Health, University of Alabama at Birmingham School of Public Health, Birmingham, AL, USA
| | - James E Hixson
- Department of Epidemiology, School of Public Health, UTHealth Houston, Houston, TX, USA
| | - Yuk-Lam Ho
- Veterans Affairs Boston Healthcare System, Boston, MA, USA
| | - Brian D Hobbs
- Department of Medicine, Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Donglei Hu
- Department of Medicine, Lung Biology Center, University of California, San Francisco, San Francisco, CA, USA
| | - Qin Hui
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
- Atlanta VA Health Care System, Decatur, GA, USA
| | - Chii-Min Hwu
- Department of Medicine, Division of Endocrinology and Metabolism, Taipei Veterans General Hospital, Taipei, Taiwan, Taiwan
| | | | - Deepti Jain
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Rita R Kalyani
- Department of Medicine, Endocrinology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sharon L R Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Tanika N Kelly
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Ethan M Lange
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Michael LeNoir
- Department of Pediatrics, Bay Area Pediatrics, Oakland, CA, USA
| | - Changwei Li
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Loic Le Marchand
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Merry-Lynn N McDonald
- Department of Medicine, Pulmonary, Allergy and Critical Care, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Caitlin P McHugh
- Department of Biostatistics, School of Public Health, University of Washington, Seattle, WA, USA
| | - Alanna C Morrison
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Take Naseri
- Naseri & Associates Public Health Consultancy Firm and Family Health Clinic, Apia, Samoa
- International Health Institute, Brown University, Providence, RI, USA
| | - Jeffrey O'Connell
- Department of Medicine, Program for Personalized and Genomic Medicine, University of Maryland, Baltimore, MD, USA
| | - Christopher J O'Donnell
- Veterans Affairs Boston Healthcare System, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Nicholette D Palmer
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - James S Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - James A Perry
- Department of Medicine, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Ulrike Peters
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Michael H Preuss
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - D C Rao
- Center for Biostatistics and Data Science, Washington University in St. Louis, St. Louis, MO, USA
| | - Elizabeth A Regan
- Department of Medicine, Rheumatology, National Jewish Health, Denver, CO, USA
| | | | - Dan M Roden
- Medicine, Pharmacology, and Biomedical Informatics, Clinical Pharmacology and Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | | | - Colleen M Sitlani
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Jennifer A Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Hemant K Tiwari
- Department of Biostatistics, University of Alabama at Birmingham School of Public Health, Birmingham, AL, USA
| | | | - Zeyuan Wang
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
| | - Daniel E Weeks
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Biostatistics and Health Data Science, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jennifer Wessel
- Department of Epidemiology, Indiana University, Indianapolis, IN, USA
- Department of Medicine, Indiana University, Indianapolis, IN, USA
- Diabaetes Translational Research Center, Indiana University, Indianapolis, IN, USA
| | - Kerri L Wiggins
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Lynne R Wilkens
- Epidemiology Program, University of Hawaii Cancer Center, Honolulu, HI, USA
| | - Peter W F Wilson
- Atlanta VA Health Care System, Decatur, GA, USA
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Lisa R Yanek
- Department of Medicine, General Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Zachary T Yoneda
- Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI, USA
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI, USA
| | - Sebastian Zöllner
- Department of Biostatistics, Department of Psychiatry, University of Michigan, Ann Arbor, MI, USA
| | - Donna K Arnett
- Department of Epidemiology, Arnold School of Public Health, University of South Carolina, Columbia, SC, USA
| | - Allison E Ashley-Koch
- Duke Molecular Physiology Institute, Duke University Medical Center, Durham, NC, USA
| | - Kathleen C Barnes
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - John Blangero
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Esteban G Burchard
- Bioengineering and Therapeutic Sciences and Medicine, Lung Biology Center, University of California, San Francisco, San Francisco, CA, USA
| | - April P Carson
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Daniel I Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Yii-Der Ida Chen
- Department of Medical Genetics, Genomic Outcomes, Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Joanne E Curran
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville, TX, USA
| | - Myriam Fornage
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Victor R Gordeuk
- Department of Medicine, School of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Jiang He
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
| | - Susan R Heckbert
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
| | - Lifang Hou
- Northwestern University, Chicago, IL, USA
| | - Marguerite R Irvin
- Department of Epidemiology, University of Alabama at Birmingham School of Public Health, Birmingham, AL, USA
| | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Ryan L Minster
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Braxton D Mitchell
- Department of Medicine, Division of Endocrinology, Diabetes and Nutrition, University of Maryland, Baltimore, MD, USA
| | - Mehdi Nouraie
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Bruce M Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Epidemiology, University of Washington, Seattle, WA, USA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA, USA
| | - Laura M Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Stephen S Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Jerome I Rotter
- Department of Pediatrics, Genomic Outcomes, The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - M Benjamin Shoemaker
- Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Nicholas L Smith
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, WA, USA
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA, USA
- Seattle Epidemiologic Research and Information Center, Office of Research and Development, Department of Veterans Affairs, Seattle, WA, USA
| | - Kent D Taylor
- Department of Pediatrics, Genomic Outcomes, The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Marilyn J Telen
- Department of Medicine, Division of Hematology, Duke University School of Medical, Durham, NC, USA
| | - Scott T Weiss
- Department of Medicine, Channing Division of Network Medicine, Harvard Medical School, Boston, MA, USA
| | - Yingze Zhang
- Department of Medicine, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nancy Heard-Costa
- Framingham Heart Study, School of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Yan V Sun
- Department of Epidemiology, Emory University Rollins School of Public Health, Atlanta, GA, USA
- Atlanta VA Health Care System, Decatur, GA, USA
| | - Xihong Lin
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Statistics, Harvard University, Cambridge, MA, USA
| | - L Adrienne Cupples
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Leslie A Lange
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Ching-Ti Liu
- Department of Biostatistics, School of Public Health, Boston University, Boston, MA, USA
| | - Ruth J F Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Anne E Justice
- Population Health Sciences, Geisinger, Danville, PA, USA.
| |
Collapse
|
23
|
Polini B, Ricardi C, Di Lupo F, Runfola M, Bacci A, Rapposelli S, Bizzarri R, Scalese M, Saponaro F, Chiellini G. Novel Thyroid Hormone Receptor-β Agonist TG68 Exerts Anti-Inflammatory, Lipid-Lowering and Anxiolytic Effects in a High-Fat Diet (HFD) Mouse Model of Obesity. Cells 2025; 14:580. [PMID: 40277905 PMCID: PMC12026167 DOI: 10.3390/cells14080580] [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: 02/19/2025] [Revised: 04/04/2025] [Accepted: 04/10/2025] [Indexed: 04/26/2025] Open
Abstract
Recent advances in drug development allowed for the identification of THRβ-selective thyromimetic TG68 as a very promising lipid lowering and anti-amyloid agent. In the current study, we first investigated the neuroprotective effects of TG68 on in vitro human models of neuroinflammation and β-amyloid neurotoxicity in order to expand our knowledge of the therapeutic potential of this novel thyromimetic. Subsequently, we examined metabolic and inflammatory profiles, along with cognitive changes, using a high-fat diet (HFD) mouse model of obesity. Our data demonstrated that TG68 was able to prevent either LPS/TNFα-induced inflammatory response or β-amyloid-induced cytotoxicity in human microglial (HMC3) cells. Next, we demonstrated that in HFD-fed mice, treatment with TG68 (10 mg/kg/day; 2 weeks) significantly reduced anxiety-like behavior in stretch-attend posture (SAP) tests while producing a 12% BW loss and a significant decrease in blood glucose and lipid levels. Notably, these data highlight a close relationship between improved serum metabolic parameters and a reduction of anxious behavior. Moreover, TG68 administration was observed to efficiently counteract HFD-altered central and peripheral expressions in mice with selected biomarkers of metabolic dysfunction, inflammation, and neurotoxicity, revealing promising neuroprotective effects. In conclusion, our work provides preliminary evidence that TG68 may represent a novel therapeutic opportunity for the treatment of interlinked diseases such as obesity and neurodegenerative diseases.
Collapse
Affiliation(s)
- Beatrice Polini
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy; (B.P.); (C.R.); (F.D.L.); (R.B.)
| | - Caterina Ricardi
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy; (B.P.); (C.R.); (F.D.L.); (R.B.)
| | - Francesca Di Lupo
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy; (B.P.); (C.R.); (F.D.L.); (R.B.)
| | - Massimiliano Runfola
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (M.R.); (A.B.); (S.R.)
| | - Andrea Bacci
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (M.R.); (A.B.); (S.R.)
| | - Simona Rapposelli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (M.R.); (A.B.); (S.R.)
| | - Ranieri Bizzarri
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy; (B.P.); (C.R.); (F.D.L.); (R.B.)
| | - Marco Scalese
- Institute of Clinical Physiology, Italian National Research Council, 56124 Pisa, Italy;
| | - Federica Saponaro
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy; (B.P.); (C.R.); (F.D.L.); (R.B.)
| | - Grazia Chiellini
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Via Roma 56, 56126 Pisa, Italy; (B.P.); (C.R.); (F.D.L.); (R.B.)
| |
Collapse
|
24
|
Downie CG, Shrestha P, Okello S, Yaser M, Lee HH, Wang Y, Krishnan M, Chen HH, Justice AE, Chittoor G, Josyula NS, Gahagan S, Blanco E, Burrows R, Correa-Burrows P, Albala C, Santos JL, Angel B, Lozoff B, Hartwig FP, Horta B, Brina KR, Isasi CR, Qi Q, Gallo LC, Perreira KM, Thyagarajan B, Daviglus M, Van Horn L, Gonzalez F, Bradfield JP, Hakonarson H, Grant SFA, Below JE, Felix J, Graff M, Divaris K, North KE. Trans-ancestry genome-wide association study of childhood body mass index identifies novel loci and age-specific effects. HGG ADVANCES 2025; 6:100411. [PMID: 39885687 PMCID: PMC11875162 DOI: 10.1016/j.xhgg.2025.100411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 01/25/2025] [Accepted: 01/25/2025] [Indexed: 02/01/2025] Open
Abstract
Over the past 30 years, obesity prevalence has markedly increased globally, including among children. Although genome-wide association studies (GWASs) have identified over 1,000 genetic loci associated with obesity-related traits in adults, the genetic architecture of childhood obesity is less well characterized. Moreover, most childhood obesity GWASs have been restricted to severely obese children, in relatively small sample sizes, and in primarily European-ancestry populations. To identify genetic loci associated with early-childhood body mass index (BMI), we performed GWAS of BMI Z scores in eight ancestrally diverse cohorts: ZOE 2.0 cohort, the Santiago Longitudinal Study (SLS), the Vanderbilt University BioVU biobank, the Geisinger MyCode Health Initiative biobank, Study of Latino (SOL) Youth, Pelotas (Brazil) Birth Cohort, Cameron County Hispanic Cohort (CCHC), and Viva La Familia cohort. We subsequently performed inverse-variance-weighted fixed-effect meta-analysis of these results with previously published GWAS summary statistics of BMI Z scores of children in the Early Growth Genetics (EGG) Consortium and the Norwegian Mother and Child Cohort (MoBa), constituting a final total of 84,804 individuals. We identified 39 genome-wide significant loci associated with childhood BMI, including three putatively novel loci (EFNA5 and DTWD2, RP11-2N5.1 on chromosome 5, and LSM14A on chromosome 19). We also observed a dynamic nature of genetic loci-BMI associations across the life course, with distinct effects across childhood and adulthood, highlighting possible critical periods for early-childhood interventions. These findings strengthen calls for larger population-based studies of children across age strata and across diverse populations.
Collapse
Affiliation(s)
- Carolina G Downie
- Department of Epidemiology, UNC Chapel Hill, Chapel Hill, NC 27514, USA.
| | - Poojan Shrestha
- Department of Epidemiology, UNC Chapel Hill, Chapel Hill, NC 27514, USA; Division of Pediatric and Public Health, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Samson Okello
- Department of Epidemiology, UNC Chapel Hill, Chapel Hill, NC 27514, USA
| | - Mohammad Yaser
- Department of Epidemiology, UNC Chapel Hill, Chapel Hill, NC 27514, USA
| | - Harold H Lee
- Department of Biobehavioral Health, Pennsylvania State University, University Park, PA 16802, USA
| | - Yujie Wang
- Department of Epidemiology, UNC Chapel Hill, Chapel Hill, NC 27514, USA
| | - Mohanraj Krishnan
- Department of Epidemiology, UNC Chapel Hill, Chapel Hill, NC 27514, USA; Carolina Population Center, UNC Chapel Hill, Chapel Hill, NC, USA
| | - Hung-Hsin Chen
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Anne E Justice
- Department of Population Health Sciences, Geisinger, Danville, PA 17822, USA
| | - Geetha Chittoor
- Department of Population Health Sciences, Geisinger, Danville, PA 17822, USA
| | | | - Sheila Gahagan
- Department of Pediatrics, University of San Diego, La Jolla, CA 92093, USA
| | - Estela Blanco
- Centro de Investigación en Sociedad y Salud y Núcleo Milenio de Sociomedicina, Universidad Mayor, Santiago, Chile
| | - Raquel Burrows
- Centro de Investigación en Sociedad y Salud y Núcleo Milenio de Sociomedicina, Universidad Mayor, Santiago, Chile
| | - Paulina Correa-Burrows
- Centro de Investigación en Sociedad y Salud y Núcleo Milenio de Sociomedicina, Universidad Mayor, Santiago, Chile
| | - Cecilia Albala
- Centro de Investigación en Sociedad y Salud y Núcleo Milenio de Sociomedicina, Universidad Mayor, Santiago, Chile
| | - José L Santos
- Department of Nutrition, Diabetes and Metabolism. School of Medicine. Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Bárbara Angel
- Public Nutrition Unit, Institute of Nutrition and Food Technology, University of Chile, Santiago, Chile
| | - Betsy Lozoff
- Department of Pediatrics, Medical School, and Department of Environmental Health Sciences, School of Public Health, University of Michigan, Ann Arbor, MI, USA
| | | | - Bernardo Horta
- Postgraduate Program in Epidemiology, Federal University of Pelotas, Pelotas, Brazil
| | - Karisa Roxo Brina
- Postgraduate Program in Epidemiology, Federal University of Pelotas, Pelotas, Brazil
| | - Carmen R Isasi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Linda C Gallo
- Department of Psychology, San Diego State University, Chula Vista, CA 91910, USA
| | - Krista M Perreira
- Department of Social Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Bharat Thyagarajan
- Department of Epidemiology, University of Minnesota Medical Center, Minneapolis, MN 55454, USA
| | - Martha Daviglus
- Department of Preventive Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Linda Van Horn
- Department of Preventive Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Franklyn Gonzalez
- Collaborative Studies Coordinating Center, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | | | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Struan F A Grant
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, PA, USA; Division of Human Genetics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Center for Spatial and Functional Genomics, The Children's Hospital of Philadelphia, Philadelphia, PA, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA; Division Endocrinology and Diabetes, The Children's Hospital of Philadelphia, Philadelphia, PA, USA; Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jennifer E Below
- Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Janine Felix
- Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands; Department of Pediatrics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Mariaelisa Graff
- Department of Epidemiology, UNC Chapel Hill, Chapel Hill, NC 27514, USA
| | - Kimon Divaris
- Department of Epidemiology, UNC Chapel Hill, Chapel Hill, NC 27514, USA; Division of Pediatric and Public Health, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kari E North
- Department of Epidemiology, UNC Chapel Hill, Chapel Hill, NC 27514, USA.
| |
Collapse
|
25
|
Huang M, Zhang W, Dong J, Hu Z, Tan X, Li H, Sun K, Zhao A, Huang T. Genome-Wide Association Studies of Body Weight and Average Daily Gain in Chinese Dongliao Black Pigs. Int J Mol Sci 2025; 26:3453. [PMID: 40244387 PMCID: PMC11989284 DOI: 10.3390/ijms26073453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 03/24/2025] [Accepted: 04/05/2025] [Indexed: 04/18/2025] Open
Abstract
In the domain of swine production, body weight (BW) and average daily gain (ADG) are recognized as the primary performance indicators. Nevertheless, the genetic architecture of ADG and BW in Dongliao black (DLB) pigs remains to be fully elucidated. In this study, we performed a genome-wide association analysis of BW, ADG, and body mass index (BMI) in 358 DLB pigs of different days of age. The genome-wide association study (GWAS) showed the following: (1) The most significant single nucleotide polymorphism (SNP) detected for BW was on Sus scrofa chromosome (SSC) 11:100,808 (p-value = 1.16 × 10-6) that was also the most significant SNP for ADG. (2) The most significant SNP associated with BMI was SSC17:51,463,521 (p-value = 5.16 × 10-8). (3) SNPs SSC10:6,523,844 and SSC17:23,852,682 were identified in both BW and ADG. A meta-analysis was conducted on BW at different days and demonstrated SSC5:39,028,335 (p-value = 8.37 × 10-6) which was not identified in the results of each single trait. The regions of two SNPs (SSC11:100,808, SSC4:10,703,277) exhibited considerable influence on both BW and ADG and the related regions were selected for linkage disequilibrium (LD) analyses that exhibited a notable linkage. In addition, several genes were identified that are associated with obesity and play roles in lipid metabolism, including MACROD2, PHLPP2, CYP2E1, and STT3B.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ayong Zhao
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou 311300, China; (M.H.); (W.Z.); (J.D.); (Z.H.); (X.T.); (H.L.); (K.S.)
| | - Tao Huang
- College of Animal Science and Technology & College of Veterinary Medicine, Zhejiang A&F University, Hangzhou 311300, China; (M.H.); (W.Z.); (J.D.); (Z.H.); (X.T.); (H.L.); (K.S.)
| |
Collapse
|
26
|
Yu H, Wang J, Xu X, Li H, Guo J. Revealing the mediating mechanisms between BMI and osteoarthritis: a Mendelian randomization and mediation analysis. Aging Clin Exp Res 2025; 37:119. [PMID: 40192902 PMCID: PMC11976339 DOI: 10.1007/s40520-025-03035-2] [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: 02/18/2025] [Accepted: 03/28/2025] [Indexed: 04/10/2025]
Abstract
BACKGROUND Despite well-documented associations between Body mass index (BMI) and Osteoarthritis (OA), the specific biological pathways and mediators involved remain poorly understood. This study aims to explore mediators through which BMI influences OA risk, particularly knee osteoarthritis (KOA), using Mendelian Randomization (MR) and mediation analysis. METHODS We used a two-step MR approach with data from the IEU OpenGWAS and FinnGen version 7 databases. BMI (N = 322,154) was the primary exposure, with knee disorders (KD), total bone mineral density (TBMD), metabolic disorders (MD), and anxiety disorders (AD) as potential mediators. Outcomes included KOA (N = 22,347), hip OA (HOA) (N = 11,989), and all OA (AllOA) (N = 50,508). Univariate MR evaluated causal relationships, followed by multivariate MR to quantify mediation effects. Multiple sensitivity analyses were conducted to validate robustness, while horizontal pleiotropy and heterogeneity were assessed using MR-Egger intercept and Cochran's Q statistic. RESULTS BMI significantly increased the risk of KOA (odds ratio [OR]: 2.00, 95% confidence interval [CI]: 1.56-2.56), HOA (OR: 2.05, 95% CI: 1.40-2.98), and AllOA (OR: 1.66, 95% CI: 1.41-1.95). KD and TBMD significantly mediated the effect on KOA, with mediation proportions of 20.89% and 3.59%, respectively. MD and AD showed no significant effects. Sensitivity analyses supported the robustness of these findings. Horizontal pleiotropy and heterogeneity tests indicated minimal evidence of bias, supporting the reliability of our results. CONCLUSIONS BMI increases OA risk, with KD and TBMD partially mediating the effect, particularly for KOA. The direct impact of BMI remains predominant, emphasizing the importance of weight reduction, joint protection, and physical activity as preventive measures.
Collapse
Affiliation(s)
- Hui Yu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, No. 555, Youyi East Road, Beilin District, Xi'an, Shaanxi, China
- Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, No. 555, Youyi East Road, Beilin District, Xi'an, Shaanxi, China
| | - Junxiang Wang
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, No. 555, Youyi East Road, Beilin District, Xi'an, Shaanxi, China
- Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, No. 555, Youyi East Road, Beilin District, Xi'an, Shaanxi, China
| | - Xin Xu
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, No. 555, Youyi East Road, Beilin District, Xi'an, Shaanxi, China
- Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, No. 555, Youyi East Road, Beilin District, Xi'an, Shaanxi, China
| | - Hui Li
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, No. 555, Youyi East Road, Beilin District, Xi'an, Shaanxi, China.
| | - Junfei Guo
- Department of Joint Surgery, Honghui Hospital, Xi'an Jiaotong University, No. 555, Youyi East Road, Beilin District, Xi'an, Shaanxi, China.
- Xi'an Key Laboratory of Pathogenesis and Precision Treatment of Arthritis, No. 555, Youyi East Road, Beilin District, Xi'an, Shaanxi, China.
| |
Collapse
|
27
|
Kianersi S, Potts KS, Wang H, Sofer T, Noordam R, Rutter MK, Redline S, Huang T. Association between accelerometer-measured irregular sleep duration and longitudinal changes in body mass index in older adults. Int J Obes (Lond) 2025:10.1038/s41366-025-01768-8. [PMID: 40189712 DOI: 10.1038/s41366-025-01768-8] [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: 12/05/2024] [Revised: 12/05/2024] [Accepted: 03/24/2025] [Indexed: 04/25/2025]
Abstract
BACKGROUND Irregular sleep duration may disrupt circadian rhythms and contribute to metabolic, behavioral, and mood changes, potentially increasing the risk for obesity. However, quantitative data on the relationship between sleep duration irregularity and weight change are lacking. METHODS In this prospective study, we analyzed data from 10,572 participants (mean age: 63 years) in the UK Biobank who wore accelerometers for a week between 2013 and 2015 and had two body mass index (BMI; kg/m²) measurements on average 2.5 years apart. Irregular sleep duration was assessed by the within-person standard deviation (SD) of 7-night accelerometer-measured sleep duration. RESULTS Participants with sleep duration SD > 60 min versus ≤30 min had 0.24 kg/m2 (95% CI: 0.08, 0.40) higher BMI change (kg/m2), standardized to three-year intervals, and 80% (95% CI: 1.28, 2.52) higher risk for incident obesity, after adjusting for sociodemographic factors, shift work, and baseline BMI or follow-up period (p-nonlinearity <0.02 for both). These associations remained consistent after adjusting for lifestyle, comorbidities, and other sleep factors, including sleep duration. Age, sex, baseline BMI, and genetic predisposition to higher BMI (measured with a polygenic risk score) did not appear to modify the association. CONCLUSIONS Since irregular sleep duration is common, trials of interventions targeting sleep irregularity might lead to new public health strategies that tackle obesity.
Collapse
Affiliation(s)
- Sina Kianersi
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Kaitlin S Potts
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Heming Wang
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Tamar Sofer
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Division of Cardiology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Raymond Noordam
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, the Netherlands
- Health Campus the Hague/Public Health and Primary Care, Leiden University Medical Center, Leiden/The Hague, the Netherlands
| | - Martin K Rutter
- Division of Diabetes, Endocrinology and Gastroenterology, School of Medical Sciences, University of Manchester, Manchester, UK
- Diabetes, Endocrinology and Metabolism Centre, Manchester Royal Infirmary, Manchester University NHS Foundation Trust, NIHR Manchester Biomedical Research Centre, Manchester Academic Health Science Centre, Manchester, UK
| | - Susan Redline
- Division of Sleep and Circadian Disorders, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Tianyi Huang
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, Baltimore, MD, USA.
| |
Collapse
|
28
|
Gaspar-Pérez A, Granero R, Fernández-Aranda F, Rosinska M, Artero C, Ruiz-Torras S, Gearhardt AN, Demetrovics Z, Guàrdia-Olmos J, Jiménez-Murcia S. Exploring Food Addiction Across Several Behavioral Addictions: Analysis of Clinical Relevance. Nutrients 2025; 17:1279. [PMID: 40219036 PMCID: PMC11990926 DOI: 10.3390/nu17071279] [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: 02/11/2025] [Revised: 03/31/2025] [Accepted: 04/01/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND/OBJECTIVES Recently, interest in studying food addiction (FA) in the context of behavioral addictions (BAs) has increased. However, research remains limited to determine the FA prevalence among various BAs. The current study aimed to investigate FA in a clinical sample of patients seeking treatment for gaming disorder, compulsive buying-shopping disorder (CBSD), compulsive sexual behavior disorder, and the comorbid presence of multiple BAs, as well as to determine the sociodemographic characteristics, personality traits, and general psychopathology of this clinical population. In addition, we analyzed whether FA is linked to a higher mean body mass index (BMI). METHODS The sample included 209 patients (135 men and 74 women) attending a specialized behavioral addiction unit. The assessment included a semi-structured clinical interview for the diagnosis of the abovementioned BAs, in addition to self-reported psychometric assessments for FA (using the Yale Food Addiction Scale 2. 0, YFAS-2), CBSD (using the Pathological Buying Screener, PBS), general psychopathology (using the Symptom Checklist-Revised, SCL-90-R), personality traits (using the Temperament and Character Inventory-Revised, TCI-R), emotional regulation (using Difficulties in Emotion Regulation Strategies, DERS), and impulsivity (using Impulsive Behavior Scale, UPPS-P). The comparison between the groups for the clinical profile was performed using logistic regression (categorical variables) and analysis of covariance (ANCOVA), adjusted based on the patients' gender. The sociodemographic profile was based on chi-square tests for categorical variables and analysis of variance (ANOVA) for quantitative measures. RESULTS The prevalence of FA in the total sample was 22.49%. The highest prevalence of FA was observed in CBSD (31.3%), followed by gaming disorder (24.7%), and the comorbid presence of multiple BAs (14.3%). No group differences (FA+/-) were found in relation to sociodemographic variables, but the comorbidity between FA and any BA was associated more with females as well as having greater general psychopathology, greater emotional dysregulation, higher levels of impulsivity, and a higher mean BMI. CONCLUSIONS The comorbidity between FA and BA is high compared to previous studies (22.49%), and it is also associated with greater severity and dysfunctionality. Emotional distress levels were high, which suggests that the group with this comorbidity may be employing FA behaviors to cope with psychological distress. However, a better understanding of the latent mechanisms that contribute to the progression of this multifaceted comorbid clinical disorder is needed. One aspect that future studies could consider is to explore the existence of FA symptoms early and routinely in patients with BAs.
Collapse
Affiliation(s)
- Anahí Gaspar-Pérez
- Doctoral Program in Clinical and Health Psychology, University of Barcelona, 08007 Barcelona, Spain; (A.G.-P.); (S.R.-T.)
- Department of Clinical Psychology, University Hospital of Bellvitge, 08908 Barcelona, Spain; (F.F.-A.); (M.R.); (C.A.)
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neuroscience Program, Bellvitge Biomedical Research Institute (IDIBELL), 08908 Barcelona, Spain;
- Ciber Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Roser Granero
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neuroscience Program, Bellvitge Biomedical Research Institute (IDIBELL), 08908 Barcelona, Spain;
- Ciber Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Psychobiology and Methodology, Autonomous University of Barcelona, 08193 Barcelona, Spain
| | - Fernando Fernández-Aranda
- Department of Clinical Psychology, University Hospital of Bellvitge, 08908 Barcelona, Spain; (F.F.-A.); (M.R.); (C.A.)
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neuroscience Program, Bellvitge Biomedical Research Institute (IDIBELL), 08908 Barcelona, Spain;
- Ciber Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08007 Barcelona, Spain
| | - Magda Rosinska
- Department of Clinical Psychology, University Hospital of Bellvitge, 08908 Barcelona, Spain; (F.F.-A.); (M.R.); (C.A.)
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neuroscience Program, Bellvitge Biomedical Research Institute (IDIBELL), 08908 Barcelona, Spain;
- Ciber Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Cristina Artero
- Department of Clinical Psychology, University Hospital of Bellvitge, 08908 Barcelona, Spain; (F.F.-A.); (M.R.); (C.A.)
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neuroscience Program, Bellvitge Biomedical Research Institute (IDIBELL), 08908 Barcelona, Spain;
| | - Silvia Ruiz-Torras
- Doctoral Program in Clinical and Health Psychology, University of Barcelona, 08007 Barcelona, Spain; (A.G.-P.); (S.R.-T.)
- Centre for Psychological Services, University of Barcelona (UB), 08035 Barcelona, Spain
| | - Ashley N Gearhardt
- Department of Psychology, University of Michigan, Ann Arbor, MI 48109, USA;
| | - Zsolt Demetrovics
- Institute for Mental Health and Wellbeing, College of Education, Psychology and Social Work, Flinders University, Adelaide, SA 5042, Australia;
- Institute of Psychology, ELTE Eötvös Loránd University, 1053 Budapest, Hungary
- Center of Excellence in Responsible Gaming, University of Gibraltar, Gibraltar GX11 1AA, Gibraltar
| | - Joan Guàrdia-Olmos
- Facultat de Psicologia, Secció de Psicologia Quantitativa, Universitat de Barcelona, 08007 Barcelona, Spain;
- UB Institute of Complex Systems, Universitat de Barcelona, 08007 Barcelona, Spain
- Institute of Neuroscience, Universitat de Barcelona, 08007 Barcelona, Spain
| | - Susana Jiménez-Murcia
- Department of Clinical Psychology, University Hospital of Bellvitge, 08908 Barcelona, Spain; (F.F.-A.); (M.R.); (C.A.)
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neuroscience Program, Bellvitge Biomedical Research Institute (IDIBELL), 08908 Barcelona, Spain;
- Ciber Fisiopatología Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, 08007 Barcelona, Spain
| |
Collapse
|
29
|
Wang H, Liu M, Li H, Xu S. Association Between Educational Attainment and Chronic Pain: A Mediation Mendelian Randomization Study. J Pain Res 2025; 18:1793-1804. [PMID: 40196193 PMCID: PMC11974555 DOI: 10.2147/jpr.s515921] [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: 01/06/2025] [Accepted: 03/19/2025] [Indexed: 04/09/2025] Open
Abstract
Background The underlying association between educational attainment (EA) and chronic pain (CP) risk is not clear. This study aimed to investigate the causal relationship of EA with CP using Mendelian randomization (MR). Methods Single nucleotide polymorphisms (SNPs) for EA were selected from the Social Science Genetic Association Consortium (SSGAC). Inverse-variance weighted (IVW), weighted median, penalized weighted median, maximum likelihood (ML), and MR-Egger methods were used to estimate causal effects. Two sample MR analyses were undertaken to assess whether EA has a causal effect on CP. We also performed mediation analyses to estimate the mediation effects. Results A genetically predicted higher EA was associated with a decreased risk of multisite chronic pain (MCP) (odds ratio [OR] = 0.772, 95% confidence interval [CI] 0.732-0.816 per one standard deviation of longer education, P < 0.05), and the Genome-wide association studies (GWAS) data for chronic widespread pain (CWP) supported the result mentioned above. Potential mediators included body mass index (BMI) (OR = 1.176, 95% CI 1.091-1.267, P < 0.05), smoking (OR = 1.054, 95% CI 1.028-1.081, P < 0.05), and depression (OR = 1.201, 95% CI 1.147-1.258, P < 0.05) have all been proven to be causally associated with MCP. The proportions of the effects of genetically predicted EA mediated through genetically predicted BMI, smoking, and depression were 17.1%, 23.6%, and 9.2%, respectively. Conclusion Genetically predicted higher educational attainment reduces multisite chronic pain risk, partially mediated by body mass index (17.1%), smoking (23.6%), and depression (9.2%), highlighting education's protective role and its potential in chronic pain prevention strategies.
Collapse
Affiliation(s)
- Hanqi Wang
- Department of Anesthesiology and Pain Research Center, The First Hospital of Jiaxing University, Jiaxing, People’s Republic of China
| | - Mingjuan Liu
- Department of Anesthesiology and Pain Research Center, The First Hospital of Jiaxing University, Jiaxing, People’s Republic of China
| | - Hongbo Li
- Department of Anesthesiology and Pain Research Center, The First Hospital of Jiaxing University, Jiaxing, People’s Republic of China
| | - Shijie Xu
- Department of Anesthesiology and Pain Research Center, The First Hospital of Jiaxing University, Jiaxing, People’s Republic of China
| |
Collapse
|
30
|
Liu Z, Zhai G. Cardiometabolic index and major depressive disorder: Stroke and diabetes as mediators. Prog Neuropsychopharmacol Biol Psychiatry 2025; 138:111340. [PMID: 40147810 DOI: 10.1016/j.pnpbp.2025.111340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 03/08/2025] [Accepted: 03/22/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Major depressive disorder (MDD) is a severe affective disorder that is clearly linked to stroke and diabetes. This study aimed to investigate the mediating role of stroke and diabetes in the association between the cardiometabolic index (CMI) and MDD. METHODS This cross-sectional study analyzed data from 8312 participants in the National Health and Nutrition Examination Survey (NHANES, 2005-2018). MDD was diagnosed using the Patient Health Questionnaire-9 (PHQ-9 score > 10). Associations were evaluated using multivariate logistic/linear regression, stratified interaction analyses, restricted cubic spline (RCS) models for nonlinearity, and bootstrap mediation testing. RESULTS There was a robust positive correlation between the incidence of MDD [OR = 1.36 (95 % CI: 1.21-1.51)] and the PHQ-9 score [β = 0.55 (95 % CI: 0.37-0.73)], with a one-unit increase in CMI. The participants in CMIQ4 had a 64 % greater risk of stroke than did the participants in CMIQ1 [OR = 1.64 (95 % CI: 1.17-2.29)]. The forest plot shows that the results remained stable under the grouping of stroke, diabetes, race, gender, and age. Moreover, stroke and diabetes both exhibited partial mediating roles, with indirect effects accounting for 4.03 % and 5.37 % of the total effect, respectively. Through RCS analysis, a nonlinear correlation was observed between CMI and MDD and between CMI and diabetes. There is a linear relationship between stroke and MDD, and maintaining CMI levels below 0.518 may mitigate the risk of MDD. CONCLUSION Stroke and diabetes partially mediated the associations between CMI and MDD. However, additional prospective studies are warranted to scrutinize the impact of CMI on MDD.
Collapse
Affiliation(s)
- Zhenyu Liu
- Department of Cardiology, Beijing Luhe Hospital, Capital Medical University, No. 82, Xinhua South Road, Tongzhou District, Beijing 101199, China.
| | - Guangyao Zhai
- Department of Cardiology, Beijing Luhe Hospital, Capital Medical University, No. 82, Xinhua South Road, Tongzhou District, Beijing 101199, China.
| |
Collapse
|
31
|
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
|
32
|
Stuber GD, Schwitzgebel VM, Lüscher C. The neurobiology of overeating. Neuron 2025:S0896-6273(25)00182-5. [PMID: 40185087 DOI: 10.1016/j.neuron.2025.03.010] [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: 12/04/2024] [Revised: 12/13/2024] [Accepted: 03/06/2025] [Indexed: 04/07/2025]
Abstract
Food intake serves to maintain energy homeostasis; however, overeating can result in obesity, which is associated with serious health complications. In this review, we explore the intricate relationship between overeating, obesity, and the underlying neurobiological mechanisms. We review the homeostatic and hedonic feeding systems, highlighting the role of the hypothalamus and reward systems in controlling food intake and energy balance. Dysregulation in both these systems leads to overeating, as seen in genetic syndromes and environmental models affecting appetite regulation when consuming highly palatable food. The concept of "food addiction" is examined, drawing parallels to drug addiction. We discuss the cellular substrate for addiction-related behavior and current pharmacological obesity treatments-in particular, GLP-1 receptor agonists-showcasing synaptic plasticity in the context of overeating and palatable food exposure. A comprehensive model integrating insights from addiction research is proposed to guide effective interventions for maladaptive feeding behaviors. Ultimately, unraveling the neurobiological basis of overeating holds promise for addressing the pressing public health issue of obesity.
Collapse
Affiliation(s)
- Garret D Stuber
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Valerie M Schwitzgebel
- Pediatric Endocrinology and Diabetes Unit, Department of Pediatrics, Gynecology and Obstetrics, Geneva University Hospitals, 1211 Geneva, Switzerland; Institute of Genetics and Genomics (iGE3) in Geneva, University of Geneva, 1211 Geneva, Switzerland
| | - Christian Lüscher
- Institute of Genetics and Genomics (iGE3) in Geneva, University of Geneva, 1211 Geneva, Switzerland; Department of Basic Neurosciences, Medical Faculty, University of Geneva, 1211 Geneva, Switzerland; Clinic of Neurology, Department of Clinical Neurosciences, Geneva University Hospitals, 1211 Geneva, Switzerland; Synapsy Center for Mental Health Research, University of Geneva, 1211 Geneva, Switzerland.
| |
Collapse
|
33
|
Yeum D, Renier TJ, Masterson TD, Carlson DD, Ballarino GA, Lansigan RK, Loos RJ, Emond JA, Gilbert-Diamond D. Genetic associations with consumption of palatable foods in the absence of hunger in response to food cues in children. Pediatr Obes 2025; 20:e13168. [PMID: 39197865 PMCID: PMC11868456 DOI: 10.1111/ijpo.13168] [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: 12/02/2023] [Revised: 07/12/2024] [Accepted: 08/05/2024] [Indexed: 09/01/2024]
Abstract
OBJECTIVE The objective of this study is to evaluate obesity-related genetic factors in relation to excess consumption and assess if food cues modify associations. METHODS Children (9-12 years) completed a randomized crossover experiment. During two visits, children ate a preload and then snacks ad libitum while watching television, embedded with food or non-food advertisements to assess eating in the absence of hunger (EAH). Primary exposures were obesity-associated genotypes, FTO rs9939609 and MC4R rs571312, and a paediatric-specific polygenic risk score (PRS). Outcomes included consumption of all snacks (total EAH) and gummy candy only (gummy candy EAH). Linear mixed-effects models tested whether genetic exposures related to EAH outcomes. We tested for effect modification by food cues using multiplicative interaction terms. RESULTS Among 177 children, each FTO risk allele was associated with a 30% increase in gummy candy EAH (p = 0.025) in adjusted models. Food cue exposure exacerbated associations between the FTO variant with gummy candy EAH (p = 0.046). No statistically significant associations were found between MC4R and EAH. CONCLUSION The results suggest children with the FTO rs9939609 risk allele may be predisposed to excess consumption of candy and that this association may be exacerbated by food cues.
Collapse
Affiliation(s)
- Dabin Yeum
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Lebanon, NH
| | - Timothy J. Renier
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Lebanon, NH
| | - Travis D. Masterson
- Department of Department of Nutritional Sciences, College of Health and Human Development, The Pennsylvania State University, University Park, PA
| | - Delaina D. Carlson
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Lebanon, NH
| | - Grace A. Ballarino
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Lebanon, NH
| | - Reina K. Lansigan
- Department of Pediatrics, Geisel School of Medicine at Dartmouth College, Dartmouth-Hitchcock Medical Center, Lebanon, NH
| | - Ruth J.F. Loos
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty for Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jennifer A. Emond
- Department of Pediatrics, Geisel School of Medicine at Dartmouth College, Dartmouth-Hitchcock Medical Center, Lebanon, NH
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth College, Lebanon, NH
| | - Diane Gilbert-Diamond
- Department of Epidemiology, Geisel School of Medicine at Dartmouth College, Lebanon, NH
- Department of Pediatrics, Geisel School of Medicine at Dartmouth College, Dartmouth-Hitchcock Medical Center, Lebanon, NH
- Department of Medicine, Geisel School of Medicine at Dartmouth College, Dartmouth-Hitchcock Medical Center, Lebanon, NH
| |
Collapse
|
34
|
Lyu Y, Lu Q, Liu Y, Xie M, Jiang L, Li J, Wang N, Dai X, Yang Y, Jiang P, Yu Q. Causal association of obesity and chronic pain mediated by educational attainment and smoking: a mediation Mendelian randomization study. Korean J Pain 2025; 38:177-186. [PMID: 40044590 PMCID: PMC11965988 DOI: 10.3344/kjp.24331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/24/2024] [Accepted: 12/26/2024] [Indexed: 04/01/2025] Open
Abstract
Background Obesity and chronic pain are related in both directions, according to earlier observational research. This research aimed to analyze the causal association between obesity and chronic pain at the genetic level, as well as to assess whether common factors mediate this relationship. Methods This study used bidirectional two sample Mendelian randomization (MR) technique to analyze the association between obesity and chronic pain. Obesity's summary genome-wide association data were obtained from European ancestry groups, as measured by body mass index (BMI), waist-to-hip ratio, waist circumference (WC), and hip circumference (HC), genome-wide association study data for chronic pain also came from the UK population, including chronic pain at three different sites (back, hip, and headache), chronic widespread pain (CWP), and multisite chronic pain (MCP). Secondly, a two-step MR and multivariate MR investigation was performed to evaluate the mediating effects of several proposed confounders. Results The authors discovered a link between chronic pain and obesity. More specifically, a sensitivity analysis was done to confirm the associations between greater BMI, WC, and HC with an increased risk of CWP and MCP. Importantly, the intermediate MR results suggest that education levels and smoking initiation may mediate the causal relationship between BMI on CWP, with a mediation effect of 23.08% and 15.38%, respectively. Conclusions The authors' findings demonstrate that the importance of education and smoking in understanding chronic pain's pathogenesis, which is important for the primary prevention and prognosis of chronic pain.
Collapse
Affiliation(s)
- Yunshu Lyu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Qingxing Lu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Yane Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Mengtong Xie
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Lintong Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Junnan Li
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Ning Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Xianglong Dai
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Yuqi Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Peiming Jiang
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| | - Qiong Yu
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, Jilin, China
| |
Collapse
|
35
|
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
|
36
|
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
|
37
|
Goulet D, Boivin M, Gravel CA, Little J, Potter BK, Dubois L. Mediation of genetic susceptibility to obesity through eating behaviours in children. Pediatr Obes 2025; 20:e13180. [PMID: 39390328 PMCID: PMC11936709 DOI: 10.1111/ijpo.13180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/09/2024] [Accepted: 09/13/2024] [Indexed: 10/12/2024]
Abstract
BACKGROUND/OBJECTIVES Few studies have examined the putative mediating role of eating behaviours linking genetic susceptibility and body weight. The goal of this study was to investigate the extent to which two polygenic scores (PGSs) for body mass index (BMI), based on child and adult data, predicted BMI through over-eating and fussy eating across childhood. SUBJECTS/METHODS The study sample involved 692 participants from a birth cohort study. Height and weight were measured on six occasions between ages 6 and 13 years. Over-eating and fussy eating behaviours were assessed five times between ages 2 and 6 years. Longitudinal growth curve mediation analysis was used to estimate the contributions of the PGSs to BMI z-scores mediated by over-eating and fussy eating. RESULTS Both PGSs predicted BMI z-scores (PGSchild: β = 0.26, 95% CI: 0.19-0.33; PGSadult: β = 0.34, 95% CI: 0.27-0.41). Over-eating significantly mediated these associations, but this mediation decreased over time from 6 years (PGSchild: 18.0%, 95% CI: 3.1-32.9, p-value = 0.018; PGSadult: 14.2%, 95% CI: 2.8-25.5, p-value = 0.014) to 13 years (PGSchild: 11.4%, 95% CI: -0.4-23.1, p-value = 0.057; PGSadult: 6.2%, 95% CI: 0.4-12.0, p-value = 0.037). Fussy eating did not show any mediation. CONCLUSIONS Our results support the view that appetite is key to translating genetic susceptibility into changes in body weight.
Collapse
Affiliation(s)
- Danick Goulet
- School of Epidemiology and Public HealthUniversity of OttawaOttawaOntarioCanada
| | | | - Christopher A. Gravel
- School of Epidemiology and Public HealthUniversity of OttawaOttawaOntarioCanada
- Department of Epidemiology, Biostatistics and Occupational HealthMcGill UniversityMontrealQuebecCanada
- Department of Mathematics and StatisticsUniversity of OttawaOttawaOntarioCanada
| | - Julian Little
- School of Epidemiology and Public HealthUniversity of OttawaOttawaOntarioCanada
| | - Beth K. Potter
- School of Epidemiology and Public HealthUniversity of OttawaOttawaOntarioCanada
| | - Lise Dubois
- School of Epidemiology and Public HealthUniversity of OttawaOttawaOntarioCanada
| |
Collapse
|
38
|
Ni M, Zhu Y, Chen Y, Zhao S, Gao A, Lu J, Wang W, Liu R, Gu W, Hong J, Wang J. A gain-of-function variant in RICTOR predisposes to human obesity. J Genet Genomics 2025; 52:549-558. [PMID: 39984155 DOI: 10.1016/j.jgg.2025.02.002] [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: 10/25/2024] [Revised: 02/09/2025] [Accepted: 02/09/2025] [Indexed: 02/23/2025]
Abstract
mTORC1/2 play central roles as signaling hubs of cell growth and metabolism and are therapeutic targets for several diseases. However, the human genetic evidence linking mutations of mTORC1/2 to obesity remains elusive. Using whole-exome sequencing of 1944 cases with severe obesity and 2161 healthy lean controls, we identify a rare RICTOR p.I116V variant enriched in 9 unrelated cases. In Rictor null mouse embryonic fibroblasts, overexpression of the RICTOR p.I116V mutant increases phosphorylation of AKT, a canonical mTORC2 substrate, compared with wild-type RICTOR, indicating a gain-of-function change. Consistent with the human obesity phenotype, the knock-in mice carrying homogenous Rictor p.I116V variants gain more body weight under a high-fat diet. Additionally, the stromal vascular fraction cells derived from inguinal white adipose tissue of knock-in mice display an enhanced capacity for adipocyte differentiation via AKT activity. These findings demonstrate that the rare gain-of-function RICTOR p.I116V mutation activates AKT signaling, promotes adipogenesis, and contributes to obesity in humans.
Collapse
Affiliation(s)
- Mengshan Ni
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Yinmeng Zhu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Yufei Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Shaoqian Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Aibo Gao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Ruixin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China
| | - Weiqiong Gu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China.
| | - Jie Hong
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China.
| | - Jiqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai 200025, China.
| |
Collapse
|
39
|
Liu Z, Zhai G. Cardiometabolic index and major depressive disorder: Stroke and diabetes as mediators. Prog Neuropsychopharmacol Biol Psychiatry 2025; 138:111340. [DOI: https:/doi.org/10.1016/j.pnpbp.2025.111340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/31/2025]
|
40
|
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
|
41
|
Wang H, Crowther TW, Isobe K, Wang H, Tateno R, Shi W. Niche Conservatism and Community Assembly Reveal Microbial Community Divergent Succession Between Litter and Topsoil. Mol Ecol 2025; 34:e17723. [PMID: 40109239 DOI: 10.1111/mec.17723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 03/22/2025]
Abstract
Natural restoration is an effective approach for restoring degraded ecosystems, yet the successional patterns and assembly mechanisms of aboveground (litter layer) and belowground (topsoil) microbial communities remain poorly understood. We applied the niche conservatism framework to investigate niche partitioning, successional patterns and community assembly processes of microbial communities in the litter and topsoil layers during long-term vegetation restoration in southwestern China. The results showed that, during vegetation succession, the potential source communities of microbial communities in the litter layer gradually shifted from being dominated by the topsoil to being dominated by the litter. Fungal communities had a significantly higher proportion of external immigrants (> 80%) than bacteria (> 40%) and archaea (< 20%). During succession, bacterial and fungal communities in the litter and topsoil layers underwent niche differentiation, displaying a divergent succession pattern, while archaeal communities showed niche overlap, following a convergent pattern driven by stochastic processes. Additionally, the dispersal rate (m) and β-diversity turnover rate (slope) of bacterial and fungal species in the litter were significantly lower than in the topsoil, with community assembly being more influenced by deterministic processes in the litter. This study reveals that higher habitat specialisation in the litter imposes stronger filtering effects on the colonisation of most microbial groups, particularly fungal communities, highlighting the role of strategy differentiation in shaping microbial communities.
Collapse
Affiliation(s)
- Haocai Wang
- Chongqing Jinfo Mountain Karst Ecosystem National Observation and Research Station, School of Geographical Sciences, Southwest University, Chongqing, China
| | - Thomas W Crowther
- Department of Environment Systems Science, Institute of Integrative Biology, ETH Zürich, Zürich, Switzerland
| | - Kazuo Isobe
- Institute of Ecology, College of Urban and Environmental Sciences, Peking University, Beijing, China
| | - Hang Wang
- Dianchi Lake Ecosystem Observation and Research Station of Yunnan Province, Kunming, China
| | - Ryunosuke Tateno
- Filed Science Education and Research Center, Kyoto University, Kyoto, Japan
| | - Weiyu Shi
- Chongqing Jinfo Mountain Karst Ecosystem National Observation and Research Station, School of Geographical Sciences, Southwest University, Chongqing, China
| |
Collapse
|
42
|
Fusco D, Marinelli C, André M, Troiani L, Noè M, Pizzagalli F, Marnetto D, Provero P. Exploring the molecular basis of the genetic correlation between body mass index and brain morphological traits. PLoS Genet 2025; 21:e1011658. [PMID: 40209151 PMCID: PMC12048161 DOI: 10.1371/journal.pgen.1011658] [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: 05/02/2025] [Accepted: 03/17/2025] [Indexed: 04/12/2025] Open
Abstract
Several studies have demonstrated significant phenotypic and genetic correlations between body mass index (BMI) and brain morphological traits derived from structural magnetic resonance imaging (sMRI). We use the sMRI, BMI, and genetic data collected by the UK Biobank to systematically compute the genetic correlations between area, volume, and thickness measurements of hundreds of brain structures on one hand, and BMI on the other. In agreement with previous literature, we find many such measurements to have negative genetic correlation with BMI. We then dissect the molecular mechanisms underlying such correlations using brain eQTL data and summary-based Mendelian randomization, thus producing an atlas of genes whose genetically regulated expression in brain tissues is pleiotropic with brain morphology and BMI. Fine-mapping followed by colocalization analysis allows, in several cases, the identification of credible sets of variants likely to be causal for both the macroscopic phenotypes and for gene expression. In particular, epigenetic fine mapping identifies variant rs7187776 in the 5' UTR of the TUFM gene as likely to be causal of increased BMI and decreased caudate volume, possibly through the creation, by the alternate allele, of an ETS binding site leading to increased chromatin accessibility, specifically in microglial cells.
Collapse
Affiliation(s)
- Daniela Fusco
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Turin, Italy
| | - Camilla Marinelli
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Turin, Italy
| | - Mathilde André
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Turin, Italy
| | - Lucia Troiani
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Turin, Italy
| | - Martina Noè
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Turin, Italy
| | - Fabrizio Pizzagalli
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Turin, Italy
| | - Davide Marnetto
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Turin, Italy
| | - Paolo Provero
- Department of Neurosciences “Rita Levi Montalcini”, University of Turin, Turin, Italy
- Center for Omics Sciences, IRCCS Ospedale San Raffaele, Milan, Italy
| |
Collapse
|
43
|
Wei L, Ding E, Lu D, Rui Z, Shen J, Fan G. Assessing the effect of modifiable risk factors on hepatocellular carcinoma: evidence from a bidirectional Mendelian randomization analysis. Discov Oncol 2025; 16:437. [PMID: 40164825 PMCID: PMC11958933 DOI: 10.1007/s12672-025-02177-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/18/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND The pathogenesis of hepatocellular carcinoma (HCC) involves a variety of environmental risk factors, some of which have yet to be fully clarified. Using the Mendelian randomization (MR) approach, this study comprehensively investigates the causal effect of genetically predicted modifiable risk factors on HCC. METHODS Genetic variants related to the 50 risk factors that had been identified in previous research were derived from genome-wide association studies. Summary statistics for the discovery cohort and validation cohort of HCC were sourced from the FinnGen consortium and the UK Biobank, respectively. Bidirectional MR analysis and sensitivity analysis were performed to establish causative risk factors for HCC. RESULTS Through the inverse variance weighted method, the results of the discovery cohort indicated that waist circumference, nonalcoholic fatty liver disease (NAFLD), alanine aminotransferase (ALT) levels, and aspartate aminotransferase (AST) levels were significantly linked to HCC occurrence risk. Furthermore, body fat percentage, glycated hemoglobin (HbA1c), obesity class 1-3, waist-to-hip ratio, iron, ferritin, transferrin saturation, and urate had suggestive associations with HCC. The validation cohort further confirmed that NAFLD and ALT levels were strongly related to HCC. Reverse MR indicated that genetic susceptibility to HCC was connected to NAFLD and transferrin saturation. Sensitivity analyses showed that most of the findings were robust. CONCLUSION This MR study delivers evidence of the complex causal relationship between modifiable risk factors and HCC. These findings offer new insights into potential prevention and treatment strategies for HCC.
Collapse
Affiliation(s)
- Lijuan Wei
- Department of Nuclear Medicine, Tianjin First Central Hospital, Tianjin, China
| | - Enci Ding
- Department of Nuclear Medicine, Tianjin First Central Hospital, Tianjin, China
| | - Dongyan Lu
- Department of Nuclear Medicine, Tianjin First Central Hospital, Tianjin, China
| | - Zhongying Rui
- Department of Nuclear Medicine, Tianjin First Central Hospital, Tianjin, China
| | - Jie Shen
- Department of Nuclear Medicine, Tianjin First Central Hospital, Tianjin, China
| | - Guoju Fan
- Department of Vascular Surgery, The Second Hospital of Tianjin Medical University, No. 23, Pingjiang Road, Hexi District, Tianjin, 300211, China.
| |
Collapse
|
44
|
Yang S, Ni X, Jian J, Wang L, Chen Z, Liu X. Is early-life obesity associated with kidney cancer in adulthood?-insights from genetic studies. Transl Androl Urol 2025; 14:519-528. [PMID: 40226061 PMCID: PMC11986479 DOI: 10.21037/tau-24-521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 03/04/2025] [Indexed: 04/15/2025] Open
Abstract
Background Adult obesity increases the risk of kidney cancer (KIC), yet the link between early body size traits and KIC remains uncertain. This study aimed to investigate the causal relationship between early body size characteristics and KIC, including its subtypes, using Mendelian randomization (MR). Methods We utilized data from public genome-wide association study (GWAS) databases on birth weight and body mass index (BMI) across childhood, adolescence, and adulthood as exposure variables, and KIC and its subtypes as outcome variables. A two-way two-sample MR analysis was performed to explore these associations, with the inverse variance weighted (IVW) method as the primary analytical approach and sensitivity analyses to assess result stability. Results IVW analysis revealed significant associations between childhood obesity [odds ratio (OR) =1.08, 95% confidence interval (CI): 1.04-1.14, P<0.001], childhood BMI (OR =1.23, 95% CI: 1.07-1.42, P=0.003), adolescent BMI (OR =1.22, 95% CI: 1.07-1.40, P=0.003), and adult BMI (OR =1.75, 95% CI: 1.41-2.17, P<0.001) with increased risk of KIC. Similar associations were observed for clear cell renal cell carcinoma (ccRCC), with childhood obesity (OR =1.09, 95% CI: 1.02-1.15, P=0.007), childhood BMI (OR =1.33, 95% CI: 1.14-1.55, P<0.001), adolescent BMI (OR =1.24, 95% CI: 1.04-1.47, P=0.01), and adult BMI (OR =1.97, 95% CI: 1.51-2.57, P<0.001) significantly linked to higher ccRCC risk. No evidence of reverse causation was found. Conclusions This study provides MR evidence supporting a causal association between early-life obesity and KIC. Our findings suggest that reducing obesity in early life may have a potential positive impact on the prevention of KIC.
Collapse
Affiliation(s)
- Song Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xinmiao Ni
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jun Jian
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lei Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhiyuan Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
45
|
Wallis NJ, McClellan A, Mörseburg A, Kentistou KA, Jamaluddin A, Dowsett GKC, Schofield E, Morros-Nuevo A, Saeed S, Lam BYH, Sumanasekera NT, Chan J, Kumar SS, Zhang RM, Wainwright JF, Dittmann M, Lakatos G, Rainbow K, Withers D, Bounds R, Ma M, German AJ, Ladlow J, Sargan D, Froguel P, Farooqi IS, Ong KK, Yeo GSH, Tadross JA, Perry JRB, Gorvin CM, Raffan E. Canine genome-wide association study identifies DENND1B as an obesity gene in dogs and humans. Science 2025; 387:eads2145. [PMID: 40048553 DOI: 10.1126/science.ads2145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 01/10/2025] [Indexed: 03/29/2025]
Abstract
Obesity is a heritable disease, but its genetic basis is incompletely understood. Canine population history facilitates trait mapping. We performed a canine genome-wide association study for body condition score-a measure of obesity-in 241 Labrador retrievers. Using a cross-species approach, we showed that canine obesity genes are also associated with rare and common forms of obesity in humans. The lead canine association was within the gene DENN domain containing 1B (DENND1B). Each copy of the alternate allele was associated with ~7.5% greater body fat. We demonstrate a role for this gene in regulating signaling and trafficking of melanocortin 4 receptor, a critical controller of energy homeostasis. Thus, canine genetics identified obesity genes and mechanisms relevant to both dogs and humans.
Collapse
Affiliation(s)
- Natalie J Wallis
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Alyce McClellan
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Alexander Mörseburg
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Katherine A Kentistou
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Aqfan Jamaluddin
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK
| | - Georgina K C Dowsett
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Ellen Schofield
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Anna Morros-Nuevo
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Sadia Saeed
- INSERM UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille, France
- University of Lille, Lille University Hospital, Lille, France
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Brian Y H Lam
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Natasha T Sumanasekera
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Justine Chan
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Sambhavi S Kumar
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Rey M Zhang
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Jodie F Wainwright
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Marie Dittmann
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Gabriella Lakatos
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Kara Rainbow
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - David Withers
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Rebecca Bounds
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre, Cambridge, UK
| | - Marcella Ma
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Alexander J German
- Institute of Life Course and Medical Sciences and School of Veterinary Science, University of Liverpool, Neston, UK
| | - Jane Ladlow
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - David Sargan
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Philippe Froguel
- INSERM UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille, France
- University of Lille, Lille University Hospital, Lille, France
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - I Sadaf Farooqi
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre, Cambridge, UK
| | - Ken K Ong
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Giles S H Yeo
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - John A Tadross
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Histopathology and Cambridge Genomics Laboratory, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - John R B Perry
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Caroline M Gorvin
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK
| | - Eleanor Raffan
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| |
Collapse
|
46
|
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
|
47
|
Goulet D, Boivin M, Gravel C, Little J, Ouellet-Morin I, Gouin JP, Dubois L. Polygenic scores of obesity in childhood based on summary statistics from adults versus children. Can J Physiol Pharmacol 2025. [PMID: 40132211 DOI: 10.1139/cjpp-2024-0221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
The lack of polygenic scores (PGSs) developed for body mass index (BMI) in children may be problematic because the genetic architecture characterizing BMI changes throughout life. This study aims to describe the genetic susceptibility to obesity in children and to compare two PGSs based on data from adults and children and their association with BMI and discrimination of obesity. The study sample comprises 717 participants aged 4-13 years. Adult- and child-based PGSs were evaluated by examining (1) mean BMI across polygenic score risk categories, (2) the capacity to identify obesity with logistic regression, and (3) the linear association with BMI z-scores using linear regression. Increases in one standardized unit of adult-based PGS were related to a stronger increase in BMI z-score (β = 0.24-0.39) than PGS derived in children (β = 0.21-0.30). The association between obesity and the child score was higher (OR = 1.75-2.33) than that for the adult score (OR = 1.74-2.06) for the age group 4-7 years. The inverse was observed for the age group 8-13 years (ORchild 1.56-1.79 vs. ORadult 1.78-2.54). Both adult- and child-based PGSs show strong associations with BMI and risk of obesity, with the adult-based score standing out from 8 years old.
Collapse
Affiliation(s)
- Danick Goulet
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
| | - Michel Boivin
- École de psychologie, Université Laval, Québec, QC, Canada
| | - Christopher Gravel
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montréal, QC, Canada
- Department of Mathematics and Statistics, University of Ottawa, Ottawa, ON, Canada
| | - Julian Little
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
| | | | | | - Lise Dubois
- School of Epidemiology and Public Health, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
48
|
Kitamoto T, Kitamoto A. Integrative proteomic and lipidomic analysis of GNB1 and SCARB2 knockdown in human subcutaneous adipocytes. PLoS One 2025; 20:e0319163. [PMID: 40127054 PMCID: PMC11932494 DOI: 10.1371/journal.pone.0319163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 01/28/2025] [Indexed: 03/26/2025] Open
Abstract
Obesity, a global public health concern, is influenced by various factors, including genetic predispositions. Although many obesity-associated genes have been identified through genome-wide association studies (GWAS), the molecular mechanisms linking these genes to adipose tissue function remain largely unexplored. This study integrates proteomic data on adipocyte fat accumulation with GWAS data on obesity to unravel the roles of the identified key candidate genes - G protein subunit beta 1 (GNB1) and scavenger receptor class B member 2 (SCARB2) - involved in fat accumulation. We utilized RNA interference to knock down GNB1 and SCARB2 in human subcutaneous adipocytes, followed by lipidome and proteome analyses using mass spectrometry. Knockdown of these genes resulted in a reduction in lipid droplet accumulation, indicating their role in adipocyte lipid storage. Digital PCR confirmed effective gene knockdown, with GNB1 and SCARB2 mRNA levels significantly reduced. In total, the lipidomic analysis identified 96 lipid species with significant alterations. GNB1 knockdown resulted in a decrease in cholesterol esters and an increase in phosphatidylcholines, phosphatidylinositols, and ceramides. SCARB2 knockdown also led to an increase in phosphatidylcholines, with a trend towards decreased triacylglycerols. Proteomic analysis revealed significant changes in proteins involved in lipid metabolism and adipocyte function, including PLPP1 and CDH13, which were upregulated following GNB1 knockdown, and HSPA8, which was downregulated. Conversely, SCARB2 knockdown resulted in the downregulation of PLPP1 and METTL7A, and the upregulation of PLIN2, HSPA8, NPC2, and SQSTM1. Our findings highlight the significant roles of GNB1 and SCARB2 in lipid metabolism and adipocyte function, providing insights that could inform therapeutic strategies targeting these regulatory genes in obesity.
Collapse
Affiliation(s)
- Takuya Kitamoto
- Advanced Research Facilities and Services, Division of Preeminent Research Supports, Institute of Photonics Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| | - Aya Kitamoto
- Advanced Research Facilities and Services, Division of Preeminent Research Supports, Institute of Photonics Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka, Japan
| |
Collapse
|
49
|
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
|
50
|
Tang B, Lin N, Liang J, Yi G, Zhang L, Peng W, Xue C, Jiang H, Li M. Leveraging pleiotropic clustering to address high proportion correlated horizontal pleiotropy in Mendelian randomization studies. Nat Commun 2025; 16:2817. [PMID: 40118820 PMCID: PMC11928562 DOI: 10.1038/s41467-025-57912-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 03/05/2025] [Indexed: 03/24/2025] Open
Abstract
Mendelian randomization harnesses genetic variants as instrumental variables to infer causal relationships between exposures and outcomes. However, certain genetic variants can affect both the exposure and the outcome through a shared factor. This phenomenon, called correlated horizontal pleiotropy, may result in false-positive causal findings. Here, we propose a Pleiotropic Clustering framework for Mendelian randomization, PCMR. PCMR detects correlated horizontal pleiotropy and extends the zero modal pleiotropy assumption to enhance causal inference in trait pairs with correlated horizontal pleiotropic variants. Simulations show that PCMR can effectively detect correlated horizontal pleiotropy and avoid false positives in the presence of correlated horizontal pleiotropic variants, even when they constitute a high proportion of the variants connecting both traits (e.g., 30-40%). In datasets consisting of 48 exposure-common disease pairs, PCMR detects horizontal correlated pleiotropy in 7 out of the exposure-common disease pairs, and avoids detecting false positive causal links. Additionally, PCMR can facilitate the integration of biological information to exclude correlated horizontal pleiotropic variants, enhancing causal inference. We apply PCMR to study causal relationships between three common psychiatric disorders as examples.
Collapse
Affiliation(s)
- Bin Tang
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Nan Lin
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Junhao Liang
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Guorong Yi
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Liubin Zhang
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Wenjie Peng
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Chao Xue
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Hui Jiang
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China
| | - Miaoxin Li
- Department of Genetics and Biomedical Informatics, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.
- Key Laboratory of Tropical Disease Control (Sun Yat-sen University), Ministry of Education, Guangzhou, 510080, China.
- Center for Precision Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China.
| |
Collapse
|