1
|
Yi X, Liu E, Wang Y. Post-genome-wide association study dissects genetic vulnerability and risk gene expression of Sjögren's disease for cardiovascular disease. J Transl Med 2025; 23:531. [PMID: 40350475 PMCID: PMC12067732 DOI: 10.1186/s12967-025-06568-2] [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: 01/08/2025] [Accepted: 05/04/2025] [Indexed: 05/14/2025] Open
Abstract
OBJECTIVES This study aims to clarify the genetic associations between Sjögren's Disease (SD) and cardiovascular disease (CVD) outcomes, and to conduct an in-depth exploration of specific pleiotropic susceptibility genes. METHODS We performed two-sample and multivariable Mendelian randomization (MR) analysis to investigate the association between SD and the risk of ischemic heart disease (IHD) and stroke. Linkage disequilibrium score regression (LDSC) and Bayesian co-localization analyses were employed to assess the genetic associations between traits. Cross-phenotype analyses were employed to identify shared variants and genes, followed by a Transcriptome-Wide Association Study (TWAS) and Multi-marker Analysis of Genomic Annotation (MAGMA) based on Multi-Trait Analysis of GWAS (MTAG) results. To validate the pleiotropic genes, we further analyzed tissue-specific differentially expressed genes (DEGs) related to SD using RNA sequencing data. RESULTS The two-sample and multivariable MR analyses revealed that SD confers a genetic vulnerability to IHD and stroke. LDSC and co-localization analyses indicated a strong genetic linkage between SD and CVDs. Cross-phenotype analyses identified 38 and 37 pleiotropic single nucleotide polymorphisms (SNPs) for SD-Stroke and SD-IHD, respectively, primarily located within the MHC class region on 6p21.32:33 loci. Additionally, TWAS and MAGMA analyses identified pleiotropic genes located outside the MHC regions-seven associated with stroke (UHRF1BP1, SNRPC, BLK, FAM167A, ARHGAP27, C8orf12, and PLEKHM1) and two associated with IHD (UHRF1BP1 and SNRPC). Proxy variants within these genes in SD suggested an increased causal risk for stroke or IHD. Co-localization analysis further reinforced that SD and stroke share significant SNPs within the loci of FAM167A, BLK, C8orf12, SNRPC, and UHRF1BP1. DEG analysis revealed a significant up-regulation of the identified genes in SD-specific tissues. CONCLUSIONS SD appears genetically predisposed to an increased risk of CVDs. Moreover, this research not only identified pleiotropic genes shared between SD and CVDs, but also, for the first time, detected key gene expressions that elevate CVD risk in SD patients-findings that may offer promising therapeutic targets for patient management.
Collapse
Affiliation(s)
- Xinglin Yi
- Department of Respiratory and Critical Care Medicine, Southwest Hospital, Army Medical University (the Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Erxiong Liu
- Department of Rheumatology and Immunology, Southwest Hospital, Army Medical University (the Third Military Medical University), Chongqing, 400038, People's Republic of China
| | - Yong Wang
- Department of Rheumatology and Immunology, Southwest Hospital, Army Medical University (the Third Military Medical University), Chongqing, 400038, People's Republic of China.
| |
Collapse
|
2
|
Li MJ, Du YX, Lan MN, Ye LY, Xu Y, Liu Y, Wu YX, Xia HY, Zhang HY, Guo M, Yang ZL, Wu ZJ, Zheng HJ. Epigenetics-Driven Appetite System Disorders and PI3K/AKT Signaling Activation Mediate a Weight Gain Resistance Phenotype Induced by a High-Fat Diet in Male Rats. Int J Eat Disord 2025. [PMID: 40237130 DOI: 10.1002/eat.24442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 04/03/2025] [Accepted: 04/03/2025] [Indexed: 04/18/2025]
Abstract
OBJECTIVE Some patients with anorexia nervosa (AN) exhibit an aversion to high-fat food. The hypothalamus plays a crucial role in feeding behavior. This study investigated the role of epigenetic modifications in the hypothalamus of male rats exhibiting a weight gain resistance (WR) phenotype induced by a high-fat diet (HFD). METHOD Male Sprague-Dawley (SD) rats were divided into normal diet (ND) and HFD groups for 9 weeks. Rats in the HFD group were then divided into HFD-induced obese (HFO) or HFD-induced WR phenotype (HFWR) groups, and body weight and food intake were monitored for 24 days. Samples such as hypothalamus and serum were collected. RESULTS The hypothalamus of HFWR rats showed increased 5-methylcytosine (5mC) levels. Whole-genome bisulfite sequencing (WGBS) analysis revealed that the level of 5mC in the hypothalamic DNA of HFWR rats was higher than that of HFO rats. The hypothalamus of HFWR rats displayed molecular disturbances in appetite systems. Through integrated analysis of the methylome and transcriptome, we discovered that alterations in methylation levels directly influenced changes in the galanin and IGF systems. Five genes (Aurkb, Cdkn1a, Galr1, ND2, and Tf) with promoter hypermethylation may be involved in appetite system disturbances. Furthermore, HFD-induced alteration of 5mC affects PI3K/Akt signaling activation, resulting in increased neuroinflammation and apoptosis in the ventromedial nucleus of the hypothalamus (VMH) in HFWR rats. DISCUSSION Our study suggests that 5mC-mediated molecular disturbances in the hypothalamic appetite system and activation of PI3K/Akt signaling in the VMH may serve as a potential pathogenic basis for HFWR.
Collapse
Affiliation(s)
- Ming-Jie Li
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Basic Medical Sciences, Fudan University, Shanghai, China
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co. Ltd., Shanghai, China
| | - Yao-Xuan Du
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Meng-Ning Lan
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lu-Yi Ye
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yu Xu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yue Liu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yi-Xiao Wu
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hai-Yang Xia
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Hua-Yue Zhang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Min Guo
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Zi-Long Yang
- College of Food Science and Technology, Shanghai Ocean University, Shanghai, China
| | - Zheng-Jun Wu
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co. Ltd., Shanghai, China
| | - Hua-Jun Zheng
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, School of Basic Medical Sciences, Fudan University, Shanghai, China
| |
Collapse
|
3
|
Isacco L, Bailly M, Lambert C, Beraud D, Duclos M, Verney J, Boirie Y, Thivel D. Influence of body composition on energy metabolism in females with constitutional thinness. Eur J Clin Nutr 2025:10.1038/s41430-025-01598-x. [PMID: 40102615 DOI: 10.1038/s41430-025-01598-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 02/14/2025] [Accepted: 03/10/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND/OBJECTIVES Individuals with constitutional thinness are characterized by an atypical resting energy metabolism and body composition. The aim of the present study was to compare patterns of energetic changes from a fasted resting state to i) postprandial resting, ii) fasted exercise, and (iii) postprandial exercise between females with constitutional thinness and normal-weight, considering body weight and composition characteristics. SUBJECTS/METHODS Body composition (X-ray absorptiometry) and maximal aerobic capacities were assessed in 17 female adults with constitutional thinness and 17 normal-weight controls. Their energy expenditure and substrate oxidation were assessed by indirect calorimetry on four occasions: i) in a fasted resting state, ii) two hours after a calibrated meal in a resting state, iii) after a 30-min cycling exercise performed postprandially, and iv) after a 20-min walking exercise in a fasted state. RESULTS Females with constitutional thinness had significantly lower body weight (p < 0.001), absolute fat-free mass (p < 0.001) and percentage of fat mass (p = 0.04). Energy expenditure was lower for all the energetic stimuli considered (from p = 0.02 to p < 0.001) in participants with constitutional thinness compared to normal-weight controls, and differences disappeared after adjustment for body weight or composition. CONCLUSIONS The present results do not indicate any impaired energetic responses to any energetic stimuli in females with constitutional thinness. Results challenge the hypothesis of a spender energy profile in individuals with constitutional thinness, and highlight the necessity to gain insight into the role of body composition in energetic regulation in this population.
Collapse
Affiliation(s)
- Laurie Isacco
- Université Clermont Auvergne, CRNH, AME2P, F-63000, Clermont-Ferrand, France.
| | - Mélina Bailly
- Université Clermont Auvergne, CRNH, AME2P, F-63000, Clermont-Ferrand, France
| | - Céline Lambert
- Biostatistics Unit, DRCI, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - Duane Beraud
- Université Clermont Auvergne, CRNH, AME2P, F-63000, Clermont-Ferrand, France
| | - Martine Duclos
- Department of Sport Medicine and Functional Explorations, CHU Clermont-Ferrand, Clermont-Ferrand, France
- UMR 1019, INRA, Clermont-Ferrand, France
- International Research Chair Health in Motion, Clermont Auvergne University Foundation, Clermont-Ferrand, France
| | - Julien Verney
- Université Clermont Auvergne, CRNH, AME2P, F-63000, Clermont-Ferrand, France
| | - Yves Boirie
- UMR 1019, INRA, Clermont-Ferrand, France
- Department of Human Nutrition, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - David Thivel
- Université Clermont Auvergne, CRNH, AME2P, F-63000, Clermont-Ferrand, France
- International Research Chair Health in Motion, Clermont Auvergne University Foundation, Clermont-Ferrand, France
| |
Collapse
|
4
|
Perdikari A, Woods VA, Ebrahim A, Lawler K, Bounds R, Singh NI, Mehlman T(S, Riley BT, Sharma S, Morris JW, Keogh JM, Henning E, Smith M, Farooqi IS, Keedy DA. Structures of human PTP1B variants reveal allosteric sites to target for weight loss therapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.08.05.603709. [PMID: 39149290 PMCID: PMC11326154 DOI: 10.1101/2024.08.05.603709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Protein Tyrosine Phosphatase 1B (PTP1B) is a negative regulator of leptin signaling whose disruption protects against diet-induced obesity in mice. We investigated whether structural characterization of human PTP1B variant proteins might reveal allosteric sites to target for weight loss therapy. To do so, we selected 12 rare variants for functional characterization from exomes from 997 people with persistent thinness and 200,000 people from UK Biobank. Seven of 12 variants impaired PTP1B function by increasing leptin-stimulated STAT3 phosphorylation in human cells. Focusing on the variants in and near the ordered catalytic domain, we ascribed structural mechanism to their functional effects using in vitro enzyme activity assays, room-temperature X-ray crystallography, and local hydrogen-deuterium exchange mass spectrometry (HDX-MS). By combining these complementary structural biology experiments for multiple variants, we characterize an inherent allosteric network in PTP1B that differs from previously reported allosteric inhibitor-driven mechanisms mediated by catalytic loop motions. The most functionally impactful variant sites map to highly ligandable surface sites, suggesting untapped opportunities for allosteric drug design. Overall, these studies can inform the targeted design of allosteric PTP1B inhibitors for the treatment of obesity.
Collapse
Affiliation(s)
- Aliki Perdikari
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science & Addenbrooke’s Hospital; Cambridge, CB2 0QQ, UK
| | - Virgil A. Woods
- Structural Biology Initiative, CUNY Advanced Science Research Center; New York, NY 10031, USA
- PhD Program in Biochemistry, CUNY Graduate Center; New York, NY 10016, USA
| | - Ali Ebrahim
- Structural Biology Initiative, CUNY Advanced Science Research Center; New York, NY 10031, USA
| | - Katherine Lawler
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science & Addenbrooke’s Hospital; Cambridge, CB2 0QQ, UK
| | - Rebecca Bounds
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science & Addenbrooke’s Hospital; Cambridge, CB2 0QQ, UK
| | - Nathanael I. Singh
- Structural Biology Initiative, CUNY Advanced Science Research Center; New York, NY 10031, USA
| | - Tamar (Skaist) Mehlman
- Structural Biology Initiative, CUNY Advanced Science Research Center; New York, NY 10031, USA
- PhD Program in Biochemistry, CUNY Graduate Center; New York, NY 10016, USA
| | - Blake T. Riley
- Structural Biology Initiative, CUNY Advanced Science Research Center; New York, NY 10031, USA
| | - Shivani Sharma
- Structural Biology Initiative, CUNY Advanced Science Research Center; New York, NY 10031, USA
- PhD Program in Biology, CUNY Graduate Center; New York, NY 10016, USA
| | - Jackson W. Morris
- Structural Biology Initiative, CUNY Advanced Science Research Center; New York, NY 10031, USA
| | - Julia M. Keogh
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science & Addenbrooke’s Hospital; Cambridge, CB2 0QQ, UK
| | - Elana Henning
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science & Addenbrooke’s Hospital; Cambridge, CB2 0QQ, UK
| | - Miriam Smith
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science & Addenbrooke’s Hospital; Cambridge, CB2 0QQ, UK
| | - I. Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Institute of Metabolic Science & Addenbrooke’s Hospital; Cambridge, CB2 0QQ, UK
| | - Daniel A. Keedy
- Structural Biology Initiative, CUNY Advanced Science Research Center; New York, NY 10031, USA
- Department of Chemistry and Biochemistry, City College of New York; New York, NY 10031, USA
- PhD Programs in Biochemistry, Biology, and Chemistry, CUNY Graduate Center; New York, NY 10016, USA
| |
Collapse
|
5
|
Martin SS, Aday AW, Allen NB, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Bansal N, Beaton AZ, Commodore-Mensah Y, Currie ME, Elkind MSV, Fan W, Generoso G, Gibbs BB, Heard DG, Hiremath S, Johansen MC, Kazi DS, Ko D, Leppert MH, Magnani JW, Michos ED, Mussolino ME, Parikh NI, Perman SM, Rezk-Hanna M, Roth GA, Shah NS, Springer MV, St-Onge MP, Thacker EL, Urbut SM, Van Spall HGC, Voeks JH, Whelton SP, Wong ND, Wong SS, Yaffe K, Palaniappan LP. 2025 Heart Disease and Stroke Statistics: A Report of US and Global Data From the American Heart Association. Circulation 2025; 151:e41-e660. [PMID: 39866113 DOI: 10.1161/cir.0000000000001303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
BACKGROUND The American Heart Association (AHA), in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, nutrition, sleep, and obesity) and health factors (cholesterol, blood pressure, glucose control, and metabolic syndrome) that contribute to cardiovascular health. The AHA Heart Disease and Stroke Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, brain health, complications of pregnancy, kidney disease, congenital heart disease, rhythm disorders, sudden cardiac arrest, subclinical atherosclerosis, coronary heart disease, cardiomyopathy, heart failure, valvular disease, venous thromboembolism, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The AHA, through its Epidemiology and Prevention Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States and globally to provide the most current information available in the annual Statistical Update with review of published literature through the year before writing. The 2025 AHA Statistical Update is the product of a full year's worth of effort in 2024 by dedicated volunteer clinicians and scientists, committed government professionals, and AHA staff members. This year's edition includes a continued focus on health equity across several key domains and enhanced global data that reflect improved methods and incorporation of ≈3000 new data sources since last year's Statistical Update. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
6
|
Ghosh S, Bouchard C. Considerations on efforts needed to improve our understanding of the genetics of obesity. Int J Obes (Lond) 2025; 49:206-210. [PMID: 38849463 PMCID: PMC11805711 DOI: 10.1038/s41366-024-01528-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 06/09/2024]
Affiliation(s)
- Sujoy Ghosh
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA.
| | - Claude Bouchard
- Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA.
| |
Collapse
|
7
|
Aleksic V, Gazibara T, Jeremic B, Kulic L, Dotlic J, Stevanovic J, Milovanovic V, Milic M. Associations of night eating with body mass index and waist circumference among health sciences students living in a post-conflict region. Nutr Health 2025:2601060241309991. [PMID: 39828953 DOI: 10.1177/02601060241309991] [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: 01/22/2025]
Abstract
BACKGROUND Stressors associated with the ongoing ethnic tensions may be the drivers of more frequent night eating, and subsequently, a higher body mass index. AIM To examine the association of night eating with body mass index (BMI) and waist circumference among health sciences students in Northern Kosovo. METHODS A cross-sectional study was carried out at the University of Prishtina in Kosovska Mitrovica from November 2018 to March 2019. Participants completed a socio-demographic questionnaire, the Night Eating Questionnaire (NEQ), the Beck's Anxiety Inventory (BAI) and the Perceived Stress Scale (PSS). Body weight, height, and waist circumference were measured by the research team. RESULTS The study sample consisted of 534 students. The night eating score was 14.4 ± 4.5 out of 52 (prevalence of night eating syndrome was 3.0%). Overall, the average BMI was 23.7 ± 4.5 kg/m2 (27.2% of students were overweight and 7.5% were obese). Moderate level of stress as measured by the PSS was observed 61.8% and high level of stress was recorded among 5.8%. Also, 9.2% students reported moderate BAI score and 6.0% students reported severe BAI score. After adjustment for socio-demographic characteristics, health behaviors, sleep, stress and anxiety levels, no association of night eating with BMI and waist circumference was observed. CONCLUSION These findings showed that there is an absence of the association between night eating, BMI and waist circumference among students who live in a post-conflict setting. There may be other factors that mediate the relationship between food intake at night and body mass index.
Collapse
Affiliation(s)
- Vojkan Aleksic
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
- Secondary School for Nursing, Cuprija, Serbia
| | - Tatjana Gazibara
- Institute of Epidemiology, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | | | - Ljiljana Kulic
- Department of Occupational Medicine, Faculty of Medicine, University of Pristina temporarily seated in Kosovska Mitrovica, Kosovska Mitrovica, Serbia
| | - Jelena Dotlic
- Clinic for Obstetrics and Gynecology, Clinical Center of Serbia, Belgrade, Serbia
- Department of Gynaecology and Obstetrics, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jasmina Stevanovic
- Department of Epidemiology, Faculty of Medicine, University of Pristina Temporarily Seated in Kosovska Mitrovica, Kosovska Mitrovica, Serbia
| | - Vesna Milovanovic
- Department of Internal Medicine, Clinical Center of Kosovska Mitrovica, Kosovska Mitrovica, Serbia
| | - Marija Milic
- Department of Epidemiology, Faculty of Medicine, University of Pristina Temporarily Seated in Kosovska Mitrovica, Kosovska Mitrovica, Serbia
- Department of Prevention, Institute of Public Health of Serbia "Dr Milan Jovanovic Batut", Belgrade, Serbia
| |
Collapse
|
8
|
Trang KB, Pahl MC, Pippin JA, Su C, Littleton SH, Sharma P, Kulkarni NN, Ghanem LR, Terry NA, O'Brien JM, Wagley Y, Hankenson KD, Jermusyk A, Hoskins J, Amundadottir LT, Xu M, Brown K, Anderson S, Yang W, Titchenell P, Seale P, Kaestner KH, Cook L, Levings M, Zemel BS, Chesi A, Wells AD, Grant SFA. 3D genomic features across >50 diverse cell types reveal insights into the genomic architecture of childhood obesity. eLife 2025; 13:RP95411. [PMID: 39813287 PMCID: PMC11735026 DOI: 10.7554/elife.95411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025] Open
Abstract
The prevalence of childhood obesity is increasing worldwide, along with the associated common comorbidities of type 2 diabetes and cardiovascular disease in later life. Motivated by evidence for a strong genetic component, our prior genome-wide association study (GWAS) efforts for childhood obesity revealed 19 independent signals for the trait; however, the mechanism of action of these loci remains to be elucidated. To molecularly characterize these childhood obesity loci, we sought to determine the underlying causal variants and the corresponding effector genes within diverse cellular contexts. Integrating childhood obesity GWAS summary statistics with our existing 3D genomic datasets for 57 human cell types, consisting of high-resolution promoter-focused Capture-C/Hi-C, ATAC-seq, and RNA-seq, we applied stratified LD score regression and calculated the proportion of genome-wide SNP heritability attributable to cell type-specific features, revealing pancreatic alpha cell enrichment as the most statistically significant. Subsequent chromatin contact-based fine-mapping was carried out for genome-wide significant childhood obesity loci and their linkage disequilibrium proxies to implicate effector genes, yielded the most abundant number of candidate variants and target genes at the BDNF, ADCY3, TMEM18, and FTO loci in skeletal muscle myotubes and the pancreatic beta-cell line, EndoC-BH1. One novel implicated effector gene, ALKAL2 - an inflammation-responsive gene in nerve nociceptors - was observed at the key TMEM18 locus across multiple immune cell types. Interestingly, this observation was also supported through colocalization analysis using expression quantitative trait loci (eQTL) derived from the Genotype-Tissue Expression (GTEx) dataset, supporting an inflammatory and neurologic component to the pathogenesis of childhood obesity. Our comprehensive appraisal of 3D genomic datasets generated in a myriad of different cell types provides genomic insights into pediatric obesity pathogenesis.
Collapse
Affiliation(s)
- Khanh B Trang
- Center for Spatial and Functional Genomics, The Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Division of Human Genetics, The Children's Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Matthew C Pahl
- Center for Spatial and Functional Genomics, The Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Division of Human Genetics, The Children's Hospital of PhiladelphiaPhiladelphiaUnited States
| | - James A Pippin
- Center for Spatial and Functional Genomics, The Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Division of Human Genetics, The Children's Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Chun Su
- Center for Spatial and Functional Genomics, The Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Division of Human Genetics, The Children's Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Sheridan H Littleton
- Center for Spatial and Functional Genomics, The Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Division of Human Genetics, The Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Genetics, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Prabhat Sharma
- Center for Spatial and Functional Genomics, The Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Pathology, The Children's Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Nikhil N Kulkarni
- Center for Spatial and Functional Genomics, The Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Pathology, The Children's Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Louis R Ghanem
- Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Natalie A Terry
- Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of PhiladelphiaPhiladelphiaUnited States
| | - Joan M O'Brien
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Penn Medicine Center for Ophthalmic Genetics in Complex DiseasePhiladelphiaUnited States
| | - Yadav Wagley
- Department of Orthopedic Surgery University of Michigan Medical School Ann ArborAnn ArborUnited States
| | - Kurt D Hankenson
- Department of Orthopedic Surgery University of Michigan Medical School Ann ArborAnn ArborUnited States
| | - Ashley Jermusyk
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer InstituteBethesdaUnited States
| | - Jason Hoskins
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer InstituteBethesdaUnited States
| | - Laufey T Amundadottir
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer InstituteBethesdaUnited States
| | - Mai Xu
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer InstituteBethesdaUnited States
| | - Kevin Brown
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer InstituteBethesdaUnited States
| | - Stewart Anderson
- Department of Child and Adolescent Psychiatry, Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Psychiatry, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Wenli Yang
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Paul Titchenell
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Physiology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Patrick Seale
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Klaus H Kaestner
- Department of Genetics, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Laura Cook
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and ImmunityMelbourneAustralia
- Department of Critical Care, Melbourne Medical School, University of MelbourneMelbourneAustralia
- Division of Infectious Diseases, Department of Medicine, University of British ColumbiaVancouverCanada
| | - Megan Levings
- Department of Surgery, University of British ColumbiaVancouverCanada
- BC Children's Hospital Research InstituteVancouverCanada
- School of Biomedical Engineering, University of British ColumbiaVancouverCanada
| | - Babette S Zemel
- Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Pediatrics, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Alessandra Chesi
- Center for Spatial and Functional Genomics, The Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Pathology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Andrew D Wells
- Center for Spatial and Functional Genomics, The Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Pathology, The Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Pathology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Institute for Immunology, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| | - Struan FA Grant
- Center for Spatial and Functional Genomics, The Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Division of Human Genetics, The Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Department of Genetics, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Pediatrics, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Division Endocrinology and Diabetes, The Children's Hospital of PhiladelphiaPhiladelphiaUnited States
- Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of PennsylvaniaPhiladelphiaUnited States
| |
Collapse
|
9
|
Thorp JG, Gerring ZF, Reay WR, Derks EM, Grotzinger AD. Genomic network analysis characterizes genetic architecture and identifies trait-specific biology. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.03.24318432. [PMID: 39677459 PMCID: PMC11643167 DOI: 10.1101/2024.12.03.24318432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Pervasive genetic overlap across human complex traits necessitates developing multivariate methods that can parse pleiotropic and trait-specific genetic signals. Here, we introduce Genomic Network Analysis (GNA), an analytic framework that applies the principles of network modelling to estimates of genetic overlap derived from genome-wide association study (GWAS) summary statistics. The result is a genomic network that describes the conditionally independent genetic associations between traits that remain when controlling for shared signal with the broader network of traits. Graph theory metrics provide added insight by formally quantifying the most important traits in the genomic network. GNA can discover additional trait-specific pathways by incorporating gene expression or genetic variants into the network to estimate their conditional associations with each trait. Extensive simulations establish GNA is well-powered for most GWAS. Application to a diverse set of traits demonstrate that GNA yields critical insight into the genetic architecture that demarcate genetically overlapping traits at varying levels of biological granularity.
Collapse
Affiliation(s)
- Jackson G Thorp
- Department of Brain and Mental Health, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Zachary F Gerring
- Department of Brain and Mental Health, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
| | - William R Reay
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Eske M Derks
- Department of Brain and Mental Health, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Andrew D Grotzinger
- Institute for Behavioral Genetics, University of Colorado Boulder, Boulder, CO
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO
| |
Collapse
|
10
|
Springer M, Meugnier E, Schnabl K, Hof KS, Champy MF, Sorg T, Petit-Demoulière B, Germain N, Galusca B, Estour B, Vidal H, Klingenspor M, Hager J. Loss of Sult1a1 reduces body weight and increases browning of white adipose tissue. Front Endocrinol (Lausanne) 2024; 15:1448107. [PMID: 39703864 PMCID: PMC11656314 DOI: 10.3389/fendo.2024.1448107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/28/2024] [Indexed: 12/21/2024] Open
Abstract
Background and objective Overweight and obesity affects millions of individuals worldwide and consequently represents a major public health concern. Individuals living with overweight and obesity have difficulty maintaining a low body weight due to known physiological mechanisms which prevent further weight loss and drive weight regain. In contrast, mechanisms which promote low body weight maintenance receive less attention and are largely unknown. To uncover these intrinsic mechanisms, we investigated a human cohort of constitutionally thin (CT) individuals which maintain a low body weight and are resistant to weight gain despite exposure to an obesogenic environment. Methods To identify novel genes that contribute to low body weight maintenance, we performed transcriptomics on adipose tissue biopsies collected from CT and normal body weight (NBW) individuals and identified sulfotransferase 1A1 (SULT1A1) as a target for further investigation in mice. Sult1a1 knockout (KO) mice were fed a standard diet to assess the impact of Sult1a1 deletion on metabolic traits. To determine if high-fat feeding recapitulated the CT weight gain resistance phenotype, Sult1a1 KO mice were fed a high-fat diet for 13-weeks. A subset of wild-type and Sult1a1 KO mice from the standard diet were further analyzed for characterization of adipose tissue respiratory capacity. Results In comparison to NBW controls, adipose tissue from CT individuals expresses less SULT1A1. Sult1a1 KO mice weigh 10% less at the end of the study period and on a high-fat diet, Sult1a1 KO mice tended to gain less weight and had reduced fat mass at 14-weeks of age. These changes were associated with reduced fasting insulin and lessened adipose tissue inflammation and fibrosis. Subcutaneous adipose tissue from Sult1a1 KO mice on a standard chow diet had elevated leak respiration, uncoupling protein 1 (UCP1) expression and increased expression of a mitochondrial marker, VDAC, associating Sult1a1 deletion to adipose tissue browning. Conclusions Our results associate Sult1a1 deletion with a tendency for lower body weight through remodeling of white adipose tissue towards a brown phenotype. The presence of UCP1, the expression of an additional mitochondrial protein and increased respiratory capacity suggest browning of the subcutaneous adipose tissue depot of Sult1a1 KO mice.
Collapse
Affiliation(s)
- Margherita Springer
- Société des Produits Nestlé S.A., Nestlé Institute of Health Sciences, Lausanne, Switzerland
- Chair for Molecular Nutritional Medicine, Technical University of Munich (TUM) School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Emmanuelle Meugnier
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAE U1397, Université Claude Bernard Lyon 1, Institut National des Sciences Appliquées de Lyon (INSA Lyon), Oullins, France
| | - Katharina Schnabl
- Chair for Molecular Nutritional Medicine, Technical University of Munich (TUM) School of Life Sciences, Technical University of Munich, Freising, Germany
- Else Kröner Fresenius Zentrum (EKFZ) für Ernährungsmedizin, Technical University of Munich, Freising, Germany
- French National Infrastructure for Mouse Phenogenomics (PHENOMIN)-Institut Clinique de la Souris, Creation, Breeding, Phenotyping, Distribution and Archiving of Model Organisms (CELPHEDIA), National Centre for Scientific Research (CNRS), National Institute of Health and Medical Research (INSERM), Université de Strasbourg, Illkirch-Grafenstaden, France
| | | | - Marie-France Champy
- French National Infrastructure for Mouse Phenogenomics (PHENOMIN)-Institut Clinique de la Souris, Creation, Breeding, Phenotyping, Distribution and Archiving of Model Organisms (CELPHEDIA), National Centre for Scientific Research (CNRS), National Institute of Health and Medical Research (INSERM), Université de Strasbourg, Illkirch-Grafenstaden, France
| | - Tania Sorg
- French National Infrastructure for Mouse Phenogenomics (PHENOMIN)-Institut Clinique de la Souris, Creation, Breeding, Phenotyping, Distribution and Archiving of Model Organisms (CELPHEDIA), National Centre for Scientific Research (CNRS), National Institute of Health and Medical Research (INSERM), Université de Strasbourg, Illkirch-Grafenstaden, France
| | - Benoit Petit-Demoulière
- French National Infrastructure for Mouse Phenogenomics (PHENOMIN)-Institut Clinique de la Souris, Creation, Breeding, Phenotyping, Distribution and Archiving of Model Organisms (CELPHEDIA), National Centre for Scientific Research (CNRS), National Institute of Health and Medical Research (INSERM), Université de Strasbourg, Illkirch-Grafenstaden, France
| | - Natacha Germain
- Division of Endocrinology, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Etienne, France
- TAPE (Eating Disorders, Addictions & Extreme Bodyweight) Research Group, University Jean Monnet, Saint Etienne, France
| | - Bogdan Galusca
- Division of Endocrinology, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Etienne, France
- TAPE (Eating Disorders, Addictions & Extreme Bodyweight) Research Group, University Jean Monnet, Saint Etienne, France
| | - Bruno Estour
- Division of Endocrinology, Centre Hospitalier Universitaire de Saint-Étienne, Saint-Etienne, France
- TAPE (Eating Disorders, Addictions & Extreme Bodyweight) Research Group, University Jean Monnet, Saint Etienne, France
| | - Hubert Vidal
- Univ-Lyon, CarMeN Laboratory, Inserm U1060, INRAE U1397, Université Claude Bernard Lyon 1, Institut National des Sciences Appliquées de Lyon (INSA Lyon), Oullins, France
| | - Martin Klingenspor
- Chair for Molecular Nutritional Medicine, Technical University of Munich (TUM) School of Life Sciences, Technical University of Munich, Freising, Germany
- Else Kröner Fresenius Zentrum (EKFZ) für Ernährungsmedizin, Technical University of Munich, Freising, Germany
- French National Infrastructure for Mouse Phenogenomics (PHENOMIN)-Institut Clinique de la Souris, Creation, Breeding, Phenotyping, Distribution and Archiving of Model Organisms (CELPHEDIA), National Centre for Scientific Research (CNRS), National Institute of Health and Medical Research (INSERM), Université de Strasbourg, Illkirch-Grafenstaden, France
| | - Jörg Hager
- Société des Produits Nestlé S.A., Nestlé Institute of Health Sciences, Lausanne, Switzerland
| |
Collapse
|
11
|
Gao C, Meng X, Liu W, Qi Q, Yan Y. Identification of sensitive periods of weight status transition over the lifespan in Chinese population. BMC Med 2024; 22:507. [PMID: 39501260 PMCID: PMC11536718 DOI: 10.1186/s12916-024-03721-4] [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: 06/06/2024] [Accepted: 10/22/2024] [Indexed: 11/09/2024] Open
Abstract
BACKGROUND The prevalence of high body mass index (BMI) is increasing in both children and adults worldwide. However, it is unclear whether vulnerabilities to maintenance and transition of weight status vary throughout the lifespan. OBJECTIVE We aimed to characterize dynamic transitions of weight statuses across different life stages and to identify the sensitive periods for maintenance, onset, and resolution of obesity. METHODS This longitudinal study included a total of 23,179 participants aged 6-80 years with 95,994 BMI measurements from the China Health and Nutrition Survey 1989-2015. To examine the heterogeneity in transitions of weight statuses across different life stages, we divided participants into 8 sub-cohorts based on baseline ages by 10-year intervals, i.e., 6-10 years, 11-20 years, 21-30 years, 31-40 years, 41-50 years, 51-60 years, 61-70 years, and 71-80 years. We estimated the probabilities of transitioning between weight statuses at a given follow-up year by baseline age using generalized linear mixed-effects models. RESULTS The predicted prevalence of obesity decreased from 6 years, bottomed at around 20 years, increased thereafter, peaked at around 55 years, and then decreased gradually. In general, participants with underweight had lower probabilities of maintaining the same status compared to those with normal weight, overweight, or obesity for all age groups. For 10-year follow-up, individuals aged 21-30 years had the highest probabilities of transitioning from normal weight to obesity and transitioning from overweight to obesity compared to those in other age groups. Individuals aged 6-20 years had the highest probabilities of transitioning from obesity to normal weight and transitioning from overweight to normal weight. Individuals in all adult age groups had higher probabilities of maintaining obesity status than children and adolescents. CONCLUSIONS Young adulthood is the most sensitive period for obesity onset, whereas childhood and adolescence are the most sensitive periods for obesity resolution. The findings suggest the heterogeneity of susceptibilities to weight status transitions across different life stages and highlight the importance of the development of age-appropriate approaches for the prevention and intervention of obesity.
Collapse
Affiliation(s)
- Chaonan Gao
- Center for Non-Communicable Disease Management, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Xin Meng
- Center for Non-Communicable Disease Management, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Wei Liu
- Center for Non-Communicable Disease Management, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Qianjin Qi
- Center for Non-Communicable Disease Management, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Yinkun Yan
- Center for Non-Communicable Disease Management, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China.
| |
Collapse
|
12
|
Verney J, Lambert C, Isacco L, Beraud D, Boscaro A, Pereira B, Costes F, Rannou F, Julian V, Duclos M, Boirie Y, Thivel D, Bailly M. Similar maximal aerobic capacity but lower energy efficiency during low-to-moderate exercise in women with constitutional thinness: new results from the NUTRILEAN study. Eur J Appl Physiol 2024; 124:3325-3336. [PMID: 38904774 DOI: 10.1007/s00421-024-05540-0] [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: 03/29/2024] [Accepted: 06/06/2024] [Indexed: 06/22/2024]
Abstract
PURPOSE Individuals with constitutional thinness have been presented with a lower muscular energy metabolism at the cellular level but their effective aerobic capacities and exercise-related energy efficiency remains unexplored. The present study compares maximal and sub-maximal aerobic capacities between subjects with constitutional thinness and age-matched normal-weight ones. METHODS Anthropometric measures, body composition (Dual-X-ray absorptiometry), physical activity and sedentary time (GT3x actigraphs), and maximal aerobic capacities (cyclingV ˙ O 2peak test) were assessed in 18 constitutionally thin (CT-body mass index < 17.5 kg m-2) and 17 normal-weight (NW-body mass index between 20 and 25 kg m-2) women. Energy efficiency was assessed during a submaximal cycling test and a walking exercise. RESULTS CT had a lower body mass and body mass index compared to NW. Absolute peak oxygen uptake and maximal aerobic power were lower in CT subjects compared to NW (ES: - 1.63 [- 2.40; - 0.86] and - 1.32 [- 2.05; - 0.58], p < 0.001).V ˙ O 2peak related to body mass was not different between groups. Gross and net efficiency (ES: - 0.78 [- 1.48; - 0.06], p = 0.03 and ES: - 0.73 [- 1.43; - 0.01], p = 0.05) were lower in CT compared to NW during the submaximal cycling exercise. The gross energy cost of walking related to body mass was lower in subjects with CT (ES: - 1.80 [- 2.60; - 0.97, p = 0.05), with no difference for the net one. Perceived exertion was similar between groups in responses to both submaximal exercises. CONCLUSION Constitutionally thin women do not show impaired aerobic capacities at moderate to maximal intensities despite lower energy efficiency while cycling and walking at low-to-moderate intensities.
Collapse
Affiliation(s)
- Julien Verney
- Laboratory of the Metabolic Adaptations to Exercise Under Physiological and Pathological Conditions (AME2P), Clermont Auvergne University, CS 60026, 63178, Aubière Cedex, France.
| | - Céline Lambert
- Biostatistics Unit, DRCI, CHU Clermont-Ferrand, 63000, Clermont-Ferrand, France
| | - Laurie Isacco
- Laboratory of the Metabolic Adaptations to Exercise Under Physiological and Pathological Conditions (AME2P), Clermont Auvergne University, CS 60026, 63178, Aubière Cedex, France
| | - Duane Beraud
- Laboratory of the Metabolic Adaptations to Exercise Under Physiological and Pathological Conditions (AME2P), Clermont Auvergne University, CS 60026, 63178, Aubière Cedex, France
| | - Audrey Boscaro
- Laboratory of the Metabolic Adaptations to Exercise Under Physiological and Pathological Conditions (AME2P), Clermont Auvergne University, CS 60026, 63178, Aubière Cedex, France
| | - Bruno Pereira
- Biostatistics Unit, DRCI, CHU Clermont-Ferrand, 63000, Clermont-Ferrand, France
| | - Frédéric Costes
- Department of Sport Medicine and Functional Explorations, CHU Clermont-Ferrand, CRNH, INRA, University of Clermont Auvergne, 63000, Clermont-Ferrand, France
| | - Fabrice Rannou
- Department of Sport Medicine and Functional Explorations, CHU Clermont-Ferrand, CRNH, INRA, University of Clermont Auvergne, 63000, Clermont-Ferrand, France
| | - Valérie Julian
- Department of Sport Medicine and Functional Explorations, CHU Clermont-Ferrand, CRNH, INRA, University of Clermont Auvergne, 63000, Clermont-Ferrand, France
| | - Martine Duclos
- Department of Sport Medicine and Functional Explorations, CHU Clermont-Ferrand, CRNH, INRA, University of Clermont Auvergne, 63000, Clermont-Ferrand, France
| | - Yves Boirie
- Department of Clinical Nutrition, CHU Clermont-Ferrand, Diet and Musculoskeletal Health Team, CRNH, INRA, University of Clermont Auvergne, 63000, Clermont-Ferrand, France
| | - David Thivel
- Laboratory of the Metabolic Adaptations to Exercise Under Physiological and Pathological Conditions (AME2P), Clermont Auvergne University, CS 60026, 63178, Aubière Cedex, France
| | - Mélina Bailly
- Laboratory of the Metabolic Adaptations to Exercise Under Physiological and Pathological Conditions (AME2P), Clermont Auvergne University, CS 60026, 63178, Aubière Cedex, France
| |
Collapse
|
13
|
Bailly M, Beraud D, Lambert C, Garnier YM, Pereira B, Duclos M, Boirie Y, Isacco L, Thivel D, Verney J. Constitutional thinness might be characterized by physiologically adapted and not impaired muscle function and architecture: new results from the NUTRILEAN study. Eur J Appl Physiol 2024; 124:3303-3315. [PMID: 38900200 DOI: 10.1007/s00421-024-05539-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/10/2024] [Indexed: 06/21/2024]
Abstract
PURPOSE While muscle mass and skeletal muscle fibers phenotype have been shown atypical in constitutional thinness (CT), force production capacities and its architectural determinants have never been explored. The present study compared muscle functionality and architecture between participants with CT and their normal-weight (NW) counterparts. METHODS Anthropometry, body composition (Dual-X-ray Absorptiometry), physical activity/sedentary behavior (ActiGraph wGT3X-BT), ultrasound recording of the Vastus Lateralis (2D-ultrasound system), and functional capacities at maximal isometric and isokinetic voluntary contractions (MVCISO and MVCCON) during knee extension (isokinetic dynamometer chair Biodex) have been measured in 18 women with CT (body mass index < 17.5 kg/m2) and 17 NW women. RESULTS A lower fat-free mass (ES: -1.94, 95%CI: -2.76 to -1.11, p < 0.001), a higher sedentary time, and a trend for a lower time spent at low-intensity physical activity, were observed in CT vs NW participants. While absolute MVCISO, MVCCON, rate of torque development (RTD), and torque work were all markedly lower in CT, these differences disappeared when normalized to body or muscle mass. Muscle thickness and fascicle length were found lower in CT (ES: -1.29, 95%CI: -2.03 to -0.52, p < 0.001; and ES: -0.87, 95%CI: -1.58 to -0.15, p = 0.02, respectively), while pennation angle was found similar. CONCLUSION Despite lower absolute strength capacities observed in CT, present findings support the hypothesis of physiological adaptations to the low body and muscle mass than to some intrinsic contractile impairments. These results call for further studies exploring hypertrophy-targeted strategies in the management of CT.
Collapse
Affiliation(s)
- Mélina Bailly
- Laboratory of the Metabolic Adaptations to Exercise Under Physiological and Pathological Conditions (AME2P), Université Clermont Auvergne, CRNH, 63000, Clermont-Ferrand, France
| | - Duane Beraud
- Laboratory of the Metabolic Adaptations to Exercise Under Physiological and Pathological Conditions (AME2P), Université Clermont Auvergne, CRNH, 63000, Clermont-Ferrand, France
| | - Céline Lambert
- Biostatistics Unit, DRCI, CHU Clermont-Ferrand, 63000, Clermont-Ferrand, France
| | - Yoann M Garnier
- Prognostic Factors and Regulatory Factors of Cardiac and Vascular Pathologies, EA3920, Université de Franche-Comté, 25000, Besançon, France
| | - Bruno Pereira
- Biostatistics Unit, DRCI, CHU Clermont-Ferrand, 63000, Clermont-Ferrand, France
| | - Martine Duclos
- Department of Sport Medicine and Functional Explorations, CHU Clermont-Ferrand, CRNH, INRA, University of Clermont Auvergne, 63000, Clermont-Ferrand, France
| | - Yves Boirie
- Department of Clinical Nutrition, CHU Clermont-Ferrand, Diet and Musculoskeletal Health Team, CRNH, INRA, University of Clermont Auvergne, 63000, Clermont-Ferrand, France
| | - Laurie Isacco
- Laboratory of the Metabolic Adaptations to Exercise Under Physiological and Pathological Conditions (AME2P), Université Clermont Auvergne, CRNH, 63000, Clermont-Ferrand, France
| | - David Thivel
- Laboratory of the Metabolic Adaptations to Exercise Under Physiological and Pathological Conditions (AME2P), Université Clermont Auvergne, CRNH, 63000, Clermont-Ferrand, France
| | - Julien Verney
- Laboratory of the Metabolic Adaptations to Exercise Under Physiological and Pathological Conditions (AME2P), Université Clermont Auvergne, CRNH, 63000, Clermont-Ferrand, France.
| |
Collapse
|
14
|
Yang Y, Sheng YH, Carreira P, Wang T, Zhao H, Wang R. Genome-wide assessment of shared genetic landscape of idiopathic pulmonary fibrosis and its comorbidities. Hum Genet 2024; 143:1223-1239. [PMID: 39103522 PMCID: PMC11485074 DOI: 10.1007/s00439-024-02696-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 07/27/2024] [Indexed: 08/07/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial lung disease accompanied by both local and systemic comorbidities. Genetic factors play a role in the development of IPF and certain associated comorbidities. Nevertheless, it is uncertain whether there are shared genetic factors underlying IPF and these comorbidities. To bridge this knowledge gap, we conducted a systematic investigation into the shared genetic architecture between IPF and ten prevalent heritable comorbidities (i.e., body mass index [BMI], coronary artery disease [CAD], chronic obstructive pulmonary disease [COPD], gastroesophageal reflux disease, lung cancer, major depressive disorder [MDD], obstructive sleep apnoea, pulmonary hypertension [PH], stroke, and type 2 diabetes), by utilizing large-scale summary data from their respective genome-wide association studies and multi-omics studies. We revealed significant (false discovery rate [FDR] < 0.05) and moderate genetic correlations between IPF and seven comorbidities, excluding lung cancer, MDD and PH. Evidence suggested a partially putative causal effect of IPF on CAD. Notably, we observed FDR-significant genetic enrichments in lung for the cross-trait between IPF and CAD and in liver for the cross-trait between IPF and COPD. Additionally, we identified 65 FDR-significant genes over-represented in 20 biological pathways related to the etiology of IPF, BMI, and COPD, including inflammation-related mucin gene clusters. Several of these genes were associated with clinically relevant drugs for the treatment of IPF, CAD, and/or COPD. Our results underscore the pervasive shared genetic basis between IPF and its common comorbidities and hold future implications for early diagnosis of IPF-related comorbidities, drug repurposing, and the development of novel therapies for IPF.
Collapse
Affiliation(s)
- Yuanhao Yang
- Mater Research Institute, The University of Queensland, Woolloongabba, QLD, Australia.
| | - Yong H Sheng
- Mater Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
- Cancer Program, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Patricia Carreira
- Immunology and Infectious Disease Division, John Curtin School of Medical Research, Australian National University, Acton, ACT, Australia
| | - Tong Wang
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Huiying Zhao
- Department of Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ran Wang
- Mater Research Institute, The University of Queensland, Woolloongabba, QLD, Australia.
| |
Collapse
|
15
|
Sharma T, Morassut RE, Langlois C, Meyre D. Body mass index trajectories and their predictors in undergraduate students from Canada: Results from the GENEiUS study. JOURNAL OF AMERICAN COLLEGE HEALTH : J OF ACH 2024; 72:2147-2155. [PMID: 35930409 DOI: 10.1080/07448481.2022.2103384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 06/29/2022] [Accepted: 07/12/2022] [Indexed: 06/15/2023]
Abstract
Objective: To explore the patterns and predictors of body mass index (BMI) change among undergraduate students from Ontario (Canada). Participants: 68 undergraduate students were followed longitudinally for 3 years with anthropometric data collected bi-annually. Methods: BMI measurements were plotted to generate individual BMI trajectory curves, which were categorized, based on the observed trajectory pattern. Within and between group comparisons of BMI were conducted via nonparametric paired tests. The association of baseline BMI, sex, and ethnicity with BMI trajectory type was assessed using multinomial logistic regression. Results: Four BMI trajectory types were observed: "stable weight" (n = 15, 22.1%), "weight gain" (n = 30, 44.1%), "weight loss" (n = 12, 17.6%), and "weight cycling" (n = 11, 16.2%) trajectories. Higher baseline BMI was significantly associated with the "weight gain," "weight loss," and the "weight cycling" trajectories as compared to the "stable weight" trajectory type. Conclusions: Our findings demonstrate an association between high baseline BMI and "nonstable" subsequent BMI change patterns among Canadian students.
Collapse
Affiliation(s)
- Tanmay Sharma
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| | - Rita E Morassut
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| | - Christine Langlois
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| | - David Meyre
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
- Department of Molecular Medicine, Division of Biochemistry, Molecular Biology, and Nutrition, University Hospital of Nancy, Nancy, France
- Faculty of Medicine of Nancy INSERM UMR_S 1256, Nutrition, Genetics, and Environmental Risk Exposure, University of Lorraine, Nancy, France
| |
Collapse
|
16
|
Trang KB, Pahl MC, Pippin JA, Su C, Littleton SH, Sharma P, Kulkarni NN, Ghanem LR, Terry NA, O’Brien JM, Wagley Y, Hankenson KD, Jermusyk A, Hoskins JW, Amundadottir LT, Xu M, Brown KM, Anderson SA, Yang W, Titchenell PM, Seale P, Cook L, Levings MK, Zemel BS, Chesi A, Wells AD, Grant SF. 3D genomic features across >50 diverse cell types reveal insights into the genomic architecture of childhood obesity. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2023.08.30.23294092. [PMID: 37693606 PMCID: PMC10491377 DOI: 10.1101/2023.08.30.23294092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
The prevalence of childhood obesity is increasing worldwide, along with the associated common comorbidities of type 2 diabetes and cardiovascular disease in later life. Motivated by evidence for a strong genetic component, our prior genome-wide association study (GWAS) efforts for childhood obesity revealed 19 independent signals for the trait; however, the mechanism of action of these loci remains to be elucidated. To molecularly characterize these childhood obesity loci we sought to determine the underlying causal variants and the corresponding effector genes within diverse cellular contexts. Integrating childhood obesity GWAS summary statistics with our existing 3D genomic datasets for 57 human cell types, consisting of high-resolution promoter-focused Capture-C/Hi-C, ATAC-seq, and RNA-seq, we applied stratified LD score regression and calculated the proportion of genome-wide SNP heritability attributable to cell type-specific features, revealing pancreatic alpha cell enrichment as the most statistically significant. Subsequent chromatin contact-based fine-mapping was carried out for genome-wide significant childhood obesity loci and their linkage disequilibrium proxies to implicate effector genes, yielded the most abundant number of candidate variants and target genes at the BDNF, ADCY3, TMEM18 and FTO loci in skeletal muscle myotubes and the pancreatic beta-cell line, EndoC-BH1. One novel implicated effector gene, ALKAL2 - an inflammation-responsive gene in nerve nociceptors - was observed at the key TMEM18 locus across multiple immune cell types. Interestingly, this observation was also supported through colocalization analysis using expression quantitative trait loci (eQTL) derived from the Genotype-Tissue Expression (GTEx) dataset, supporting an inflammatory and neurologic component to the pathogenesis of childhood obesity. Our comprehensive appraisal of 3D genomic datasets generated in a myriad of different cell types provides genomic insights into pediatric obesity pathogenesis.
Collapse
Affiliation(s)
- Khanh B. Trang
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Matthew C. Pahl
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - James A. Pippin
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Chun Su
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Sheridan H. Littleton
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Prabhat Sharma
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nikhil N. Kulkarni
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Louis R. Ghanem
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, PA, USA
| | - Natalie A. Terry
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, PA, USA
| | - Joan M. O’Brien
- Scheie Eye Institute, Department of Ophthalmology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, PA, USA
- Penn Medicine Center for Ophthalmic Genetics in Complex Disease
| | - Yadav Wagley
- Department of Orthopedic Surgery University of Michigan Medical School Ann Arbor, MI, USA
| | - Kurt D. Hankenson
- Department of Orthopedic Surgery University of Michigan Medical School Ann Arbor, MI, USA
| | - Ashley Jermusyk
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Jason W. Hoskins
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Laufey T. Amundadottir
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Mai Xu
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Kevin M Brown
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Stewart A. Anderson
- Department of Child and Adolescent Psychiatry, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wenli Yang
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul M. Titchenell
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Patrick Seale
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Laura Cook
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Department of Critical Care, Melbourne Medical School, University of Melbourne, Melbourne, VIC, Australia
- Division of Infectious Diseases, Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Megan K. Levings
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
- BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Babette S. Zemel
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Alessandra Chesi
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew D. Wells
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pathology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Struan F.A. Grant
- Center for Spatial and Functional Genomics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Division of Human Genetics, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Institute for Diabetes, Obesity and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division Endocrinology and Diabetes, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Penn Neurodegeneration Genomics Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
17
|
Farooqi IS, Xu Y. Translational potential of mouse models of human metabolic disease. Cell 2024; 187:4129-4143. [PMID: 39067442 DOI: 10.1016/j.cell.2024.07.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/05/2024] [Accepted: 07/05/2024] [Indexed: 07/30/2024]
Abstract
Obesity causes significant morbidity and mortality globally. Research in the last three decades has delivered a step-change in our understanding of the fundamental mechanisms that regulate energy homeostasis, building on foundational discoveries in mouse models of metabolic disease. However, not all findings made in rodents have translated to humans, hampering drug discovery in this field. Here, we review how studies in mice and humans have informed our current framework for understanding energy homeostasis, discuss their challenges and limitations, and offer a perspective on how human studies may play an increasingly important role in the discovery of disease mechanisms and identification of therapeutic targets in the future.
Collapse
Affiliation(s)
- I Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, Cambridge, UK.
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Department of Molecular and Cellular Biology and Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
18
|
AlMarzooqi SK, Almarzooqi F, Sadida HQ, Jerobin J, Ahmed I, Abou-Samra AB, Fakhro KA, Dhawan P, Bhat AA, Al-Shabeeb Akil AS. Deciphering the complex interplay of obesity, epithelial barrier dysfunction, and tight junction remodeling: Unraveling potential therapeutic avenues. Obes Rev 2024; 25:e13766. [PMID: 38745386 DOI: 10.1111/obr.13766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 03/11/2024] [Accepted: 04/17/2024] [Indexed: 05/16/2024]
Abstract
Obesity stands as a formidable global health challenge, predisposing individuals to a plethora of chronic illnesses such as cardiovascular disease, diabetes, and cancer. A confluence of genetic polymorphisms, suboptimal dietary choices, and sedentary lifestyles significantly contribute to the elevated incidence of obesity. This multifaceted health issue profoundly disrupts homeostatic equilibrium at both organismal and cellular levels, with marked alterations in gut permeability as a salient consequence. The intricate mechanisms underlying these alterations have yet to be fully elucidated. Still, evidence suggests that heightened inflammatory cytokine levels and the remodeling of tight junction (TJ) proteins, particularly claudins, play a pivotal role in the manifestation of epithelial barrier dysfunction in obesity. Strategic targeting of proteins implicated in these pathways and metabolites such as short-chain fatty acids presents a promising intervention for restoring barrier functionality among individuals with obesity. Nonetheless, recognizing the heterogeneity among affected individuals is paramount; personalized medical interventions or dietary regimens tailored to specific genetic backgrounds and allergy profiles may prove indispensable. This comprehensive review delves into the nexus of obesity, tight junction remodeling, and barrier dysfunction, offering a critical appraisal of potential therapeutic interventions.
Collapse
Affiliation(s)
- Sara K AlMarzooqi
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| | - Fajr Almarzooqi
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| | - Hana Q Sadida
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| | - Jayakumar Jerobin
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Ikhlak Ahmed
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| | - Abdul-Badi Abou-Samra
- Qatar Metabolic Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar
| | - Khalid A Fakhro
- Department of Human Genetics, Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
- Department of Genetic Medicine, Weill Cornell Medicine, Doha, Qatar
| | - Punita Dhawan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, Nebraska, USA
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| | - Ammira S Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Research Program, Sidra Medicine, Doha, Qatar
| |
Collapse
|
19
|
Powell DR, Doree DD, Shadoan MK, Platt KA, Brommage R, Vogel P, Revelli JP. Mice Lacking Mrs2 Magnesium Transporter are Hypophagic and Thin When Maintained on a High-Fat Diet. Endocrinology 2024; 165:bqae072. [PMID: 38878275 DOI: 10.1210/endocr/bqae072] [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: 03/06/2024] [Indexed: 07/05/2024]
Abstract
Genes regulating body fat are shared with high fidelity by mice and humans, indicating that mouse knockout (KO) phenotyping might identify valuable antiobesity drug targets. Male Mrs2 magnesium transporter (Mrs2) KO mice were recently reported as thin when fed a high-fat diet (HFD). They also exhibited increased energy expenditure (EE)/body weight and had beiged adipocytes that, along with isolated hepatocytes, demonstrated increased oxygen consumption, suggesting that increased EE drove the thin phenotype. Here we provide our data on these and additional assays in Mrs2 KO mice. We generated Mrs2 KO mice by homologous recombination. HFD-fed male and female Mrs2 KO mice had significantly less body fat, measured by quantitative magnetic resonance, than wild-type (WT) littermates. HFD-fed Mrs2 KO mice did not demonstrate increased EE by indirect calorimetry and could not maintain body temperature at 4 °C, consistent with their decreased brown adipose tissue stores but despite increased beige white adipose tissue. Instead, when provided a choice between HFD and low-fat diet (LFD), Mrs2 KO mice showed a significant 15% decrease in total energy intake resulting from significantly lower HFD intake that offset numerically increased LFD intake. Food restriction studies performed using WT mice suggested that this decrease in energy intake could explain the loss of body fat. Oral glucose tolerance test studies revealed significantly improved insulin sensitivity in Mrs2 KO mice. We conclude that HFD-fed Mrs2 KO mice are thin with improved insulin sensitivity, and that this favorable metabolic phenotype is driven by hypophagia. Further evaluation is warranted to determine the suitability of MRS2 as a drug target for antiobesity therapeutics.
Collapse
Affiliation(s)
| | - Deon D Doree
- Lexicon Pharmaceuticals, The Woodlands, TX 77381, USA
| | | | | | | | - Peter Vogel
- Lexicon Pharmaceuticals, The Woodlands, TX 77381, USA
| | | |
Collapse
|
20
|
Martin SS, Aday AW, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Barone Gibbs B, Beaton AZ, Boehme AK, Commodore-Mensah Y, Currie ME, Elkind MSV, Evenson KR, Generoso G, Heard DG, Hiremath S, Johansen MC, Kalani R, Kazi DS, Ko D, Liu J, Magnani JW, Michos ED, Mussolino ME, Navaneethan SD, Parikh NI, Perman SM, Poudel R, Rezk-Hanna M, Roth GA, Shah NS, St-Onge MP, Thacker EL, Tsao CW, Urbut SM, Van Spall HGC, Voeks JH, Wang NY, Wong ND, Wong SS, Yaffe K, Palaniappan LP. 2024 Heart Disease and Stroke Statistics: A Report of US and Global Data From the American Heart Association. Circulation 2024; 149:e347-e913. [PMID: 38264914 DOI: 10.1161/cir.0000000000001209] [Citation(s) in RCA: 845] [Impact Index Per Article: 845.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
BACKGROUND The American Heart Association (AHA), in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, nutrition, sleep, and obesity) and health factors (cholesterol, blood pressure, glucose control, and metabolic syndrome) that contribute to cardiovascular health. The AHA Heart Disease and Stroke Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, brain health, complications of pregnancy, kidney disease, congenital heart disease, rhythm disorders, sudden cardiac arrest, subclinical atherosclerosis, coronary heart disease, cardiomyopathy, heart failure, valvular disease, venous thromboembolism, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The AHA, through its Epidemiology and Prevention Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States and globally to provide the most current information available in the annual Statistical Update with review of published literature through the year before writing. The 2024 AHA Statistical Update is the product of a full year's worth of effort in 2023 by dedicated volunteer clinicians and scientists, committed government professionals, and AHA staff members. The AHA strives to further understand and help heal health problems inflicted by structural racism, a public health crisis that can significantly damage physical and mental health and perpetuate disparities in access to health care, education, income, housing, and several other factors vital to healthy lives. This year's edition includes additional global data, as well as data on the monitoring and benefits of cardiovascular health in the population, with an enhanced focus on health equity across several key domains. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
21
|
Gkouskou KK, Grammatikopoulou MG, Lazou E, Vasilogiannakopoulou T, Sanoudou D, Eliopoulos AG. A genomics perspective of personalized prevention and management of obesity. Hum Genomics 2024; 18:4. [PMID: 38281958 PMCID: PMC10823690 DOI: 10.1186/s40246-024-00570-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 01/03/2024] [Indexed: 01/30/2024] Open
Abstract
This review discusses the landscape of personalized prevention and management of obesity from a nutrigenetics perspective. Focusing on macronutrient tailoring, we discuss the impact of genetic variation on responses to carbohydrate, lipid, protein, and fiber consumption. Our bioinformatic analysis of genomic variants guiding macronutrient intake revealed enrichment of pathways associated with circadian rhythm, melatonin metabolism, cholesterol and lipoprotein remodeling and PPAR signaling as potential targets of macronutrients for the management of obesity in relevant genetic backgrounds. Notably, our data-based in silico predictions suggest the potential of repurposing the SYK inhibitor fostamatinib for obesity treatment in relevant genetic profiles. In addition to dietary considerations, we address genetic variations guiding lifestyle changes in weight management, including exercise and chrononutrition. Finally, we emphasize the need for a refined understanding and expanded research into the complex genetic landscape underlying obesity and its management.
Collapse
Affiliation(s)
- Kalliopi K Gkouskou
- Department of Biology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, 11527, Athens, Greece.
- GENOSOPHY P.C., Athens, Greece.
| | - Maria G Grammatikopoulou
- Unit of Immunonutrition and Clinical Nutrition, Department of Rheumatology and Clinical Immunology, University General Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, Larissa, Greece
| | | | - Theodora Vasilogiannakopoulou
- Department of Biology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, 11527, Athens, Greece
| | - Despina Sanoudou
- Clinical Genomics and Pharmacogenomics Unit, 4th Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Aristides G Eliopoulos
- Department of Biology, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, 11527, Athens, Greece.
- GENOSOPHY P.C., Athens, Greece.
- Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece.
| |
Collapse
|
22
|
Milhem F, Skates E, Wilson M, Komarnytsky S. Obesity-Resistant Mice on a High-Fat Diet Display a Distinct Phenotype Linked to Enhanced Lipid Metabolism. Nutrients 2024; 16:171. [PMID: 38202000 PMCID: PMC10780630 DOI: 10.3390/nu16010171] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 12/22/2023] [Accepted: 01/01/2024] [Indexed: 01/12/2024] Open
Abstract
Individually, metabolic variations can significantly influence predisposition to obesity in the form of the obesity-prone (super-responders) and obesity-resistant (non-responders) phenotypes in response to modern calorie-dense diets. In this study, C57BL/6J mice (n = 76) were randomly assigned to either a low-fat diet (LFD) or a high-fat diet (HFD) for 6 weeks, followed by selection of the normally obese (HFD), non-responders (NR), super-responders (SR), or super-responders switched back to the low-fat diet (SR-LFD) for an additional 8 weeks. SR mice showed the highest gains in body weight, lean and fat body mass, and total and free water, in part due to increased feed efficiency, despite having a respiratory exchange ratio (RER) similar to that of NR mice. A switch to the LFD was sufficient to revert most of the observed physiological changes in the SR-LFD mice; however, voluntary physical activity and exercise capacity did not return to the basal level. NR mice showed the highest food intake, lowest feed efficiency, increased oxygen consumption during the light (rest) cycle, increased physical activity during the dark (active) cycle, and increased heat production during both cycles. These variations were observed in the absence of changes in food intake and fecal parameters; however, NR fecal lipid content was lower, and the NR fecal microbiome profile was characterized by reduced abundance of Actinobacteria. Taken together, our findings suggest that NR mice showed an increased ability to metabolize excessive dietary fats in skeletal muscle at the expense of reduced exercise capacity that persisted for the duration of the study. These findings underscore the need for further comprehensive investigations into the mechanisms of obesity resistance, as they hold potential implications for weight-loss strategies in human subjects.
Collapse
Affiliation(s)
- Fadia Milhem
- Plants for Human Health Institute, NC State University, 600 Laureate Way, Kannapolis, NC 28081, USA; (F.M.); (E.S.); (M.W.)
- Department of Food, Bioprocessing, and Nutrition Sciences, NC State University, 400 Dan Allen Drive, Raleigh, NC 27695, USA
- Department of Nutrition, University of Petra, 317 Airport Road, Amman 11196, Jordan
| | - Emily Skates
- Plants for Human Health Institute, NC State University, 600 Laureate Way, Kannapolis, NC 28081, USA; (F.M.); (E.S.); (M.W.)
| | - Mickey Wilson
- Plants for Human Health Institute, NC State University, 600 Laureate Way, Kannapolis, NC 28081, USA; (F.M.); (E.S.); (M.W.)
| | - Slavko Komarnytsky
- Plants for Human Health Institute, NC State University, 600 Laureate Way, Kannapolis, NC 28081, USA; (F.M.); (E.S.); (M.W.)
- Department of Food, Bioprocessing, and Nutrition Sciences, NC State University, 400 Dan Allen Drive, Raleigh, NC 27695, USA
| |
Collapse
|
23
|
Hübel C, Abdulkadir M, Herle M, Palmos AB, Loos RJF, Breen G, Micali N, Bulik CM. Persistent thinness and anorexia nervosa differ on a genomic level. Eur J Hum Genet 2024; 32:117-124. [PMID: 37474786 PMCID: PMC10772076 DOI: 10.1038/s41431-023-01431-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 06/15/2023] [Accepted: 07/04/2023] [Indexed: 07/22/2023] Open
Abstract
Thinness and anorexia nervosa are both characterised by persistent low weight. Individuals with anorexia nervosa concurrently report distorted perceptions of their body and engage in weight-loss behaviours, whereas individuals with thinness often wish to gain weight. Both conditions are heritable and share genomics with BMI, but are not genetically correlated with each other. Based on their pattern of genetic associations with other traits, we explored differences between thinness and anorexia nervosa on a genomic level. In Part 1, using publicly available data, we compared genetic correlations of persistent thinness/anorexia nervosa with eleven psychiatric disorders. In Part 2, we identified individuals with adolescent persistent thinness in the Avon Longitudinal Study of Parents and Children (ALSPAC) by latent class growth analysis of measured BMI from 10 to 24 years (n = 6594) and evaluated associations with psychiatric and anthropometric polygenic scores. In Part 1, in contrast to the positive genetic correlations of anorexia nervosa with various psychiatric disorders, persistent thinness showed negative genetic correlations with attention deficit hyperactivity disorder (rgAN = 0.08 vs. rgPT = -0.30), alcohol dependence (rgAN = 0.07 vs. rgPT = -0.44), major depressive disorder (rgAN = 0.27 vs. rgPT = -0.18) and post-traumatic stress disorder (rgAN = 0.26 vs. rgPT = -0.20). In Part 2, individuals with adolescent persistent thinness in the ALSPAC had lower borderline personality disorder polygenic scores (OR = 0.77; Q = 0.01). Overall, results suggest that genetic variants associated with thinness are negatively associated with psychiatric disorders and therefore thinness may be differentiable from anorexia nervosa on a genomic level.
Collapse
Affiliation(s)
- Christopher Hübel
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK.
- National Institute for Health Research (NIHR) Maudsley Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, UK.
- National Centre for Register-based Research, Aarhus Business and Social Sciences, Aarhus University, Aarhus, Denmark.
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
- Department of Pediatric Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
| | - Mohamed Abdulkadir
- National Centre for Register-based Research, Aarhus Business and Social Sciences, Aarhus University, Aarhus, Denmark
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Moritz Herle
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- Department of Biostatistics & Health Informatics, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Alish B Palmos
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- National Institute for Health Research (NIHR) Maudsley Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, UK
| | - Ruth J F Loos
- Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen, Denmark
| | - Gerome Breen
- Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
- National Institute for Health Research (NIHR) Maudsley Biomedical Research Centre at South London and Maudsley NHS Foundation Trust, London, UK
| | - Nadia Micali
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland
- Great Ormond Street Institute of Child Health, University College London, London, UK
- Mental Health Services in the Capital Region of Denmark, Eating Disorders Research Unit, Psychiatric Centre Ballerup, Ballerup, Denmark
| | - Cynthia M Bulik
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
24
|
Boscaro A, Verney J, Tremblay A, King JA, Pereira B, Costes F, Julian V, Duclos M, Boirie Y, Thivel D, Bailly M. Challenges of considering both extremities of the weight status spectrum to better understand obesity: insights from the NUTRILEAN project in constitutionally thin individuals. Int J Obes (Lond) 2023; 47:1171-1177. [PMID: 37553452 DOI: 10.1038/s41366-023-01360-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/03/2023] [Accepted: 07/27/2023] [Indexed: 08/10/2023]
Abstract
BACKGROUND/OBJECTIVES While the physiology of obesity has been so extensively investigated to date, only an extremely small number of studies (less than 50) have focused on the other extremity of the weight spectrum: constitutional thinness. Yet, this important state of underweight in the absence of any eating disorders provides a mirror model of obesity that might be particularly insightful in understanding obesity. Nevertheless, important methodological and recruitment-related issues appear when it comes to this complex constitutionally thin phenotype, as experienced by our research group with the realization of the ongoing NUTRILEAN clinical trial. To face this challenge, the present paper aims at identifying, analyzing, and discussing the quality of such recruitment processes in publications about constitutional thinness. METHODS In this order, a group of experts collectively created a new grading system to assess the level of rigour and quality achieved by each study based on different criteria. RESULTS The main results were that (i) metabolic-related biasing criteria were poorly observed despite being crucial, (ii) recruitment processes were not detailed enough and with sufficient explicitness, and (iii) recruiting among already identified patients would be associated with both higher sample sizes and better scores of quality. CONCLUSIONS The present work encourages investigators to adopt a high level of rigour despite the complexity and duration of recruitment processes for this specific population, and readers to pay close attention to the quality of recruitment when interpreting the data. To better understand obesity and its physiological adaptations, it seems essential not only to compare it to normal-weight conditions, but also to the other extremity of the weight status spectrum represented by constitutional thinness.
Collapse
Affiliation(s)
- Audrey Boscaro
- Laboratory of the Metabolic Adaptations to Exercise under Physiological and Pathological Conditions (AME2P), UPR 3533, CRNH Auvergne, University of Clermont Auvergne, Clermont-Ferrand, France.
| | - Julien Verney
- Laboratory of the Metabolic Adaptations to Exercise under Physiological and Pathological Conditions (AME2P), UPR 3533, CRNH Auvergne, University of Clermont Auvergne, Clermont-Ferrand, France
| | - Angelo Tremblay
- Department of Kinesiology, Faculty of Medicine, Laval University, Quebec, QC, Canada
| | - James A King
- National Centre for Sport and Exercise Medicine, School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
- NIHR Leicester Biomedical Research Centre, University Hospitals of Leicester NHS Trust and the University of Leicester, Leicester, UK
| | - Bruno Pereira
- Biostatistics Unit, Délégation à la Recherche Clinique et à l'Innovation (DRCI), Clermont-Ferrand, France
| | - Frédéric Costes
- Department of Sport Medicine and Functional Explorations, University Teaching Hospital of Clermont-Ferrand, Diet and Musculoskeletal Health Team, CRNH, INRA, University of Clermont Auvergne, Clermont-Ferrand, France
| | - Valérie Julian
- Department of Sport Medicine and Functional Explorations, University Teaching Hospital of Clermont-Ferrand, Diet and Musculoskeletal Health Team, CRNH, INRA, University of Clermont Auvergne, Clermont-Ferrand, France
| | - Martine Duclos
- Department of Sport Medicine and Functional Explorations, University Teaching Hospital of Clermont-Ferrand, Diet and Musculoskeletal Health Team, CRNH, INRA, University of Clermont Auvergne, Clermont-Ferrand, France
| | - Yves Boirie
- Department of Clinical Nutrition, University Teaching Hospital of Clermont-Ferrand, Diet and Musculoskeletal Health Team, CRNH, INRA, University of Clermont Auvergne, Clermont-Ferrand, France
| | - David Thivel
- Laboratory of the Metabolic Adaptations to Exercise under Physiological and Pathological Conditions (AME2P), UPR 3533, CRNH Auvergne, University of Clermont Auvergne, Clermont-Ferrand, France
| | - Mélina Bailly
- Laboratory of the Metabolic Adaptations to Exercise under Physiological and Pathological Conditions (AME2P), UPR 3533, CRNH Auvergne, University of Clermont Auvergne, Clermont-Ferrand, France
| |
Collapse
|
25
|
Farooqi S. Obesity and thinness: insights from genetics. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220205. [PMID: 37661743 PMCID: PMC10475868 DOI: 10.1098/rstb.2022.0205] [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: 03/15/2023] [Accepted: 06/09/2023] [Indexed: 09/05/2023] Open
Abstract
Genetic disruption of key molecular components of the hypothalamic leptin-melanocortin pathway causes severe obesity in mice and humans. Physiological studies in people who carry these mutations have shown that the adipose tissue-derived hormone leptin primarily acts to defend against starvation. A lack of leptin causes an intense drive to eat and increases the rewarding properties of food, demonstrating that human appetite has a strong biological basis. Genetic studies in clinical- and population-based cohorts of people with obesity or thinness continue to provide new insights into the physiological mechanisms involved in weight regulation and identify molecular targets for weight loss therapy. This article is part of a discussion meeting issue 'Causes of obesity: theories, conjectures and evidence (Part II)'.
Collapse
Affiliation(s)
- Sadaf Farooqi
- Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Box 289, Cambridge CB2 0QQ, UK
| |
Collapse
|
26
|
Abstract
Non-Alcoholic Fatty Liver Disease (NAFLD) refers to the accumulation of lipid laden vacuoles in hepatocytes, occurring in the context of visceral adiposity, insulin resistance and other features of the metabolic syndrome. Its more severe form (NASH, Non-Alcoholic Steatohepatitis) is becoming the leading aetiology of end-stage liver disease and hepatocellular carcinoma, and also contributes to cardiovascular disease, diabetes and extrahepatic malignancy. Management is currently limited to lifestyle modification and optimisation of the metabolic co-morbidities, with some of the drugs used for the latter also having shown some benefit for the liver. Licensed treatment modalities are currently lacking. A particular difficulty is the notorious heterogeneity of the patient population, which is poorly understood. A spectrum of disease severity associates in a non-linear way with a spectrum of severity of underlying metabolic factors. Heterogeneity of the liver in terms of mechanisms to cope with the metabolic and inflammatory stress and in terms of repair mechanisms, and a lack of knowledge hereof, further complicate the understanding of inter-individual variability. Genetic factors act as disease modifiers and potentially allow for some risk stratification, but also only explain a minor fraction of disease heterogeneity. Response to treatment shows a large variation in treatment response, again with little understanding of what is driving the absence of response in individual patients. Management can be tailored to patient's preferences in terms of diet modification, but tailoring treatment to knowledge on disease driving mechanisms in an individual patient is still in its infancy. Recent progress in analysing liver tissue as well as non-invasive tests hold, however, promise to rapidly improve our understanding of disease heterogeneity in NAFLD and provide individualised management.
Collapse
Affiliation(s)
- Sven M Francque
- Department of Gastroenterology Hepatology, Antwerp University Hospital, Drie Eikenstraat 655, B-2650, Edegem, Belgium.
- InflaMed Centre of Excellence, Laboratory for Experimental Medicine and Paediatrics, Translational Sciences in Inflammation and Immunology, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, B-2610, Wilrijk, Belgium.
| |
Collapse
|
27
|
Lund J, Clemmensen C. Physiological protection against weight gain: evidence from overfeeding studies and future directions. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220229. [PMID: 37482786 PMCID: PMC10363696 DOI: 10.1098/rstb.2022.0229] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 04/24/2023] [Indexed: 07/25/2023] Open
Abstract
Body weight is under physiological regulation. When body fat mass decreases, a series of responses are triggered to promote weight regain by increasing food intake and decreasing energy expenditure. Analogous, in response to experimental overfeeding, excessive weight gain is counteracted by a reduction in food intake and possibly by an increase in energy expenditure. While low blood leptin and other hormones defend against weight loss, the signals that oppose overfeeding-induced fat mass expansion are still unknown. In this article, we discuss insights gained from overfeeding interventions in humans and intragastric overfeeding studies in rodents. We summarize the knowledge on the relative contributions of energy intake, energy expenditure and energy excretion to the physiological defence against overfeeding-induced weight gain. Furthermore, we explore literature supporting the existence of unidentified endocrine and non-endocrine pathways that defend against weight gain. Finally, we discuss the physiological drivers of constitutional thinness and suggest that overfeeding of individuals with constitutional thinness represents a gateway to understand the physiology of weight gain resistance in humans. Experimental overfeeding, combined with modern multi-omics techniques, has the potential to unveil the long-sought signalling pathways that protect against weight gain. Discovering these mechanisms could give rise to new treatments for obesity. This article is part of a discussion meeting issue 'Causes of obesity: theories, conjectures and evidence (Part I)'.
Collapse
Affiliation(s)
- Jens Lund
- Novo Nordisk Foundation Center for Basic Metabolic Research. Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Christoffer Clemmensen
- Novo Nordisk Foundation Center for Basic Metabolic Research. Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
28
|
Sweeney P, Gimenez LE, Hernandez CC, Cone RD. Targeting the central melanocortin system for the treatment of metabolic disorders. Nat Rev Endocrinol 2023; 19:507-519. [PMID: 37365323 DOI: 10.1038/s41574-023-00855-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/18/2023] [Indexed: 06/28/2023]
Abstract
A large body of preclinical and clinical data shows that the central melanocortin system is a promising therapeutic target for treating various metabolic disorders such as obesity and cachexia, as well as anorexia nervosa. Setmelanotide, which functions by engaging the central melanocortin circuitry, was approved by the FDA in 2020 for use in certain forms of syndromic obesity. Furthermore, the FDA approvals in 2019 of two peptide drugs targeting melanocortin receptors for the treatment of generalized hypoactive sexual desire disorder (bremelanotide) and erythropoietic protoporphyria-associated phototoxicity (afamelanotide) demonstrate the safety of this class of peptides. These approvals have also renewed excitement in the development of therapeutics targeting the melanocortin system. Here, we review the anatomy and function of the melanocortin system, discuss progress and challenges in developing melanocortin receptor-based therapeutics, and outline potential metabolic and behavioural disorders that could be addressed using pharmacological agents targeting these receptors.
Collapse
Affiliation(s)
- Patrick Sweeney
- School of Molecular and Cellular Biology, College of Liberal Arts and Sciences, University of Illinois Urbana-Champaign, Champaign, IL, USA
| | - Luis E Gimenez
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
| | | | - Roger D Cone
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA.
- Department of Molecular and Integrative Physiology, School of Medicine, University of Michigan, Ann Arbor, MI, USA.
- Department of Molecular, Cellular, and Developmental Biology, College of Literature Science and the Arts, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
29
|
Stensel DJ. How can physical activity facilitate a sustainable future? Reducing obesity and chronic disease. Proc Nutr Soc 2023; 82:286-297. [PMID: 36892103 DOI: 10.1017/s0029665123002203] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
This review examines the ways in which physical activity can contribute to a sustainable future by addressing significant public health issues. The review begins by identifying obesity and ageing as two major challenges facing societies around the world due to the association of both with the risk of chronic disease. Recent developments in the understanding and treatment of obesity are examined followed by an appraisal of the role of exercise alone and in combination with other therapies in preventing and managing obesity. The review then addresses the interaction between exercise and appetite due to the central role appetite plays in the development of overweight and obesity. The final section of the review examines the potential of physical activity to combat age-related chronic disease risk including CVD, cancer and dementia. It is concluded that while bariatric surgery and pharmacotherapy are the most effective treatments for severe obesity, physical activity has a role to play facilitating and enhancing weight loss in combination with other methods. Where weight/fat reduction via exercise is less than expected this is likely due to metabolic adaptation induced by physiological changes facilitating increased energy intake and decreased energy expenditure. Physical activity has many health benefits independent of weight control including reducing the risk of developing CVD, cancer and dementia and enhancing cognitive function in older adults. Physical activity may also provide resilience for future generations by protecting against the more severe effects of global pandemics and reducing greenhouse gas emissions via active commuting.
Collapse
Affiliation(s)
- David J Stensel
- School of Sport, Exercise and Health Sciences, National Centre for Sport and Exercise Medicine, Loughborough University, Loughborough, UK
- National Institute for Health and Care Research (NIHR), Leicester Biomedical Research Centre, University Hospitals of Leicester, National Health Service (NHS) Trust and the University of Leicester, Leicester, UK
- Faculty of Sport Sciences, Waseda University, Tokorozawa, Japan
- Department of Sports Science and Physical Education, The Chinese University of Hong Kong, Ma Liu Shui, Hong Kong
| |
Collapse
|
30
|
Zhou JY, Liu M, Park S. Interaction of environmental factors with the polygenic risk scores of thinness-related genes in preventing obesity risk in middle-aged adults: The KoGES. J Hum Nutr Diet 2023; 36:1451-1467. [PMID: 36632775 DOI: 10.1111/jhn.13132] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 12/19/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND Some persons are genetically resistant to obesity, but only a few studies have evaluated thinness genes for preventing obesity. We aimed to investigate the association of polygenic variants with being underweight and their interaction with the lifestyles of middle-aged and elderly persons and identify potential new genetic approaches for managing body weight. METHODS In total, 58,701 participants aged 40-77 years were recruited from urban hospitals in Korea. Underweight (case) was defined as body mass index (BMI) < 18.5 kg m2 (n = 991) and normal weight (control, n = 21,921) was defined as 18.5 ≤ BMI < 23 kg m2 . A genome-wide association study was run to identify thinness-related single nucleotide polymorphisms (SNPs) after adjustment for compound factors using Gplink. The generalised multifactor dimensionality reduction program was used to identify the genetic variants with SNP-SNP interactions. The polygenic risk score (PRS) was calculated by summing up the number of risk alleles in each SNP and classifying them into low-, medium- and high-PRS. RESULTS The best model included the ANK2_rs7656666, CAST_rs28042, SLC1A3_rs928431867, CHST12_rs2906173, ALOX5_rs1051713, RGS6_rs17180754, ST8SIA5_rs79491311 and DCC_rs35721894 alleles. The participants with high-PRS had a lower BMI (p < 0.0001) than those with low-PRS and were 3.834 (2.58-5.70) times more likely to be underweight after multivariate adjustment (p < 0.001). The selected SNPs were correlated with each other and highly expressed in brain-related genes. The genes with minor alleles of CAST_rs28042 and CHST12_rs2906173 exhibited a higher expression frequency in brain-related tissues. PRS had significant interactions with protein, sodium, indigestible carbohydrates, calcium intake and exercise (p < 0.05), influencing the underweight state. People with a high-PRS were more underweight than those with low-PRS under high protein, sodium, high calcium, low indigestible carbohydrate intake and low exercise by 3.75, 3.88, 7.05, 3.18 and 3.80 times, respectively (p < 0.0001). CONCLUSIONS In conclusion, adults having a high-PRS were significantly correlated with being underweight, especially in combination with a particular nutritional status. These results show the potential for thinness genes to be applied to personalised nutrition for preventing obesity through targeted gene therapy.
Collapse
Affiliation(s)
- Jun-Yu Zhou
- Department of Bioconvergence System, Hoseo University, Asan, Korea
| | - Meiling Liu
- Department of Food and Nutrition, Obesity/Diabetes Research Center, Hoseo University, Asan, Korea
| | - Sunmin Park
- Department of Bioconvergence System, Hoseo University, Asan, Korea
- Department of Food and Nutrition, Obesity/Diabetes Research Center, Hoseo University, Asan, Korea
| |
Collapse
|
31
|
Becetti I, Bwenyi EL, de Araujo IE, Ard J, Cryan JF, Farooqi IS, Ferrario CR, Gluck ME, Holsen LM, Kenny PJ, Lawson EA, Lowell BB, Schur EA, Stanley TL, Tavakkoli A, Grinspoon SK, Singhal V. The Neurobiology of Eating Behavior in Obesity: Mechanisms and Therapeutic Targets: A Report from the 23rd Annual Harvard Nutrition Obesity Symposium. Am J Clin Nutr 2023; 118:314-328. [PMID: 37149092 PMCID: PMC10375463 DOI: 10.1016/j.ajcnut.2023.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 04/03/2023] [Accepted: 05/01/2023] [Indexed: 05/08/2023] Open
Abstract
Obesity is increasing at an alarming rate. The effectiveness of currently available strategies for the treatment of obesity (including pharmacologic, surgical, and behavioral interventions) is limited. Understanding the neurobiology of appetite and the important drivers of energy intake (EI) can lead to the development of more effective strategies for the prevention and treatment of obesity. Appetite regulation is complex and is influenced by genetic, social, and environmental factors. It is intricately regulated by a complex interplay of endocrine, gastrointestinal, and neural systems. Hormonal and neural signals generated in response to the energy state of the organism and the quality of food eaten are communicated by paracrine, endocrine, and gastrointestinal signals to the nervous system. The central nervous system integrates homeostatic and hedonic signals to regulate appetite. Although there has been an enormous amount of research over many decades regarding the regulation of EI and body weight, research is only now yielding potentially effective treatment strategies for obesity. The purpose of this article is to summarize the key findings presented in June 2022 at the 23rd annual Harvard Nutrition Obesity Symposium entitled "The Neurobiology of Eating Behavior in Obesity: Mechanisms and Therapeutic Targets." Findings presented at the symposium, sponsored by NIH P30 Nutrition Obesity Research Center at Harvard, enhance our current understanding of appetite biology, including innovative techniques used to assess and systematically manipulate critical hedonic processes, which will shape future research and the development of therapeutics for obesity prevention and treatment.
Collapse
Affiliation(s)
- Imen Becetti
- Division of Pediatric Endocrinology, Massachusetts General Hospital for Children and Harvard Medical School, Boston, MA, United States.
| | - Esther L Bwenyi
- Metabolism Unit, Massachusetts General Hospital, Boston, MA, United States; Nutrition Obesity Research Center at Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
| | - Ivan E de Araujo
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY, United States; Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Jamy Ard
- Epidemiology and Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Bariatric and Weight Management Center, Wake Forest Baptist Health, Winston-Salem, NC, United States; Center on Diabetes, Obesity, and Metabolism, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Sticht Center for Healthy Aging and Alzheimer's Prevention, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Hypertension and Vascular Research Center, Cardiovascular Sciences Center, Wake Forest University School of Medicine, Winston-Salem, NC, United States; Maya Angelou Center for Healthy Equity, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Ismaa Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories and National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, United Kingdom; Wellcome-Medical Research Council (MRC) Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom; Addenbrooke's Hospital, Cambridge University Hospitals, Cambridge, United Kingdom
| | - Carrie R Ferrario
- Department of Pharmacology, Psychology Department (Biopsychology Area), University of Michigan, Ann Arbor, MI, United States
| | - Marci E Gluck
- National Institutes of Health, Phoenix, AZ, United States; National Institute of Diabetes and Digestive and Kidney Disease, Obesity and Diabetes Clinical Research Section, Phoenix Epidemiology and Clinical Research Branch, Phoenix, AZ, United States
| | - Laura M Holsen
- Harvard Medical School, Boston, MA, United States; Division of Women's Health, Department of Medicine, Brigham and Women's Hospital, Boston, MA, United States; Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, United States
| | - Paul J Kenny
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY, United States; Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York City, NY, United States
| | - Elizabeth A Lawson
- Nutrition Obesity Research Center at Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States; Neuroendocrine Unit, Massachusetts General Hospital, Boston, MA, United States
| | - Bradford B Lowell
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Ellen A Schur
- Division of General Internal Medicine, University of Washington, Seattle, WA, United States; Univeristy of Washington Medicine Diabetes Institute, University of Washington, Seattle, WA, United States; Univeristy of Washington Nutrition and Obesity Research Center, University of Washington, Seattle, WA, United States; Clinical and Translational Research Services Core, University of Washington, Seattle, WA, United States
| | - Takara L Stanley
- Division of Pediatric Endocrinology, Massachusetts General Hospital for Children and Harvard Medical School, Boston, MA, United States; Metabolism Unit, Massachusetts General Hospital, Boston, MA, United States; Nutrition Obesity Research Center at Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States
| | - Ali Tavakkoli
- Division of General and Gastrointestinal (GI) Surgery, Center for Weight Management and Wellness, Advanced Minimally Invasive Fellowship, Harvard Medical School, Boston, MA, United States
| | - Steven K Grinspoon
- Metabolism Unit, Massachusetts General Hospital, Boston, MA, United States; Nutrition Obesity Research Center at Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States; Harvard Medical School, Boston, MA, United States; Department of Medicine, Harvard Medical School, Boston, MA, United States
| | - Vibha Singhal
- Division of Pediatric Endocrinology, Massachusetts General Hospital for Children and Harvard Medical School, Boston, MA, United States; Harvard Medical School, Boston, MA, United States; Pediatric Endocrinology and Obesity Medicine, Massachusetts General Hospital, Boston, MA, United States; Pediatric Program MGH Weight Center, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
32
|
O'Hara V, Cuda S, Kharofa R, Censani M, Conroy R, Browne NT. Clinical review: Guide to pharmacological management in pediatric obesity medicine. OBESITY PILLARS 2023; 6:100066. [PMID: 37990657 PMCID: PMC10661861 DOI: 10.1016/j.obpill.2023.100066] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/18/2023] [Accepted: 04/20/2023] [Indexed: 11/23/2023]
Abstract
Introduction Newer pharmacotherapy agents (anti-obesity medication [AOM]) are revolutionizing the management of children and adolescents with obesity. Previously, treatment based on intensive behavioral therapy involved many patient and family contact hours and yielded improvements in obesity status of 1-3 percent of the 95th percentile of the body mass index (BMI). Newer AOMs are yielding more clinically significant improvement of 5-18 percent. This review provides guidance for practitioners in the care of children and adolescents with obesity who frequently have complex medical and behavioral health care needs. Specifically, we discuss the use of newer AOMs in these complex patients. Methods This review details an approach to the care of the child and adolescent with obesity using AOMs. A shared decision-making process is presented in which the provider and the patient and family collaborate on care. Management of medical and behavioral components of the disease of obesity in the child are discussed. Results Early aggressive treatment is recommended, starting with an assessment of associated medical and behavioral complications, weight promoting medications, use of AOMs and ongoing care. Intensive behavioral therapy is foundational to treatment, but not a specific treatment. Patients and families deserve education on expected outcomes with each therapeutic option. Conclusions The use of new AOMs in children and adolescents has changed expected clinical outcomes in the field of pediatric obesity management. Clinically significant improvement in obesity status occurs when AOMs are used early and aggressively. Ongoing, chronic care is the model for optimizing outcomes using a shared decision-making between provider and patient/family. Depending on the experience and comfort level of the primary care practitioner, referral to an obesity medicine specialist may be appropriate, particularly when obesity related co-morbidities are present and pharmacotherapy and metabolic and bariatric surgery are considerations.
Collapse
Affiliation(s)
- Valerie O'Hara
- Weight & Wellness Clinic, Maine Medical Center, S. Portland, ME, 04106, USA
| | - Suzanne Cuda
- Alamo City Healthy Kids and Families, 1919 Oakwell Farms Parkway, Ste 145, San Antonio, TX, 78218, USA
| | - Roohi Kharofa
- Department of Pediatrics, University of Cincinnati College of Medicine, Center for Better Health & Nutrition, The Heart Institute, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229, USA
| | - Marisa Censani
- Clinical Pediatrics, Division of Pediatric Endocrinology, Department of Pediatrics, New York Presbyterian Hospital, Weill Cornell Medicine, 525 East 68th Street, Box 103, New York, NY, 10021, USA
| | - Rushika Conroy
- Division of Pediatric Endocrinology, Baystate Children's Hospital Subspecialty Center, 50 Wason Avenue, Springfield, MA, 01107, USA
| | | |
Collapse
|
33
|
Whitfield H, Hargreaves D, Nicholls D, Watt HC, Creese H. Risk factors of persistent adolescent thinness: findings from the UK Millennium Cohort Study. BMC Public Health 2023; 23:938. [PMID: 37226159 DOI: 10.1186/s12889-023-15850-1] [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: 08/02/2022] [Accepted: 05/09/2023] [Indexed: 05/26/2023] Open
Abstract
BACKGROUND Thinness during adolescence can increase the risk of adverse health outcomes across the life-course and impede development. There is limited research examining the prevalence and determinants of persistent adolescent thinness in the United Kingdom (UK). We used longitudinal cohort data to investigate determinants of persistent adolescent thinness. METHODS We analyzed data from 7,740 participants in the UK Millennium Cohort Study at ages 9 months, 7, 11, 14 and 17 years. Persistent thinness was defined as thinness at ages 11, 14 and 17; thinness was defined as an age- and sex-adjusted Body Mass Index (BMI) of less than 18.5 kg/m2. In total, 4,036 participants, classified either as persistently thin or at a persistent healthy weight, were included in the analyses. Logistic regression analyses were conducted to examine associations between 16 risk factors and persistent adolescent thinness by sex. RESULTS The prevalence of persistent thinness among adolescents was 3.1% (n = 231). Among males (n = 115), persistent adolescent thinness was significantly associated with non-white ethnicity, low parental BMI, low birthweight, low breastfeeding duration, unintended pregnancy, and low maternal education. Among females (n = 116), persistent adolescent thinness was significantly associated with non-white ethnicity, low birthweight, low self-esteem, and low physical activity. However, after adjusting for all risk factors, only low maternal BMI (OR: 3.44; 95% CI:1.13, 10.5), low paternal BMI (OR: 22.2; 95% CI: 2.35, 209.6), unintended pregnancy (OR: 2.49; 95% CI: 1.11, 5.57) and low self-esteem (OR: 6.57; 95% CI: 1.46,29.7) remained significantly associated with persistent adolescent thinness among males. After adjustment for all risk factors, not reaching the recommended physical activity levels (OR: 4.22; 95% CI: 1.82, 9.75) remained significantly associated with persistent adolescent thinness among females. No appreciable associations were found between persistent adolescent thinness and sex, premature birth, smoking during pregnancy, income, maternal postnatal depression, mother-infant attachment or socio-emotional difficulties (p > 0.05). CONCLUSION Persistent adolescent thinness is not rare and appears to be associated with both physical and mental health factors, with some sex specific differences. Healthy weight initiatives should consider the full weight spectrum. Further research is required to understand thinness at a population level, including among those whose BMI changes during child and adolescent development.
Collapse
Affiliation(s)
- H Whitfield
- School of Public Health, Imperial College London, London, UK.
| | - D Hargreaves
- School of Public Health, Imperial College London, London, UK
| | - D Nicholls
- Department of Brain Sciences, Imperial College London, London, UK
| | - H C Watt
- School of Public Health, Imperial College London, London, UK
| | - H Creese
- School of Public Health, Imperial College London, London, UK
| |
Collapse
|
34
|
Toelsie JR, Morpurgo F, Krishnadath I, Bipat R. Obesity, overweight and hyperglycemia among primary school children in a low-middle income country with a multiethnic population. OBESITY PILLARS (ONLINE) 2023; 5:100053. [PMID: 37990748 PMCID: PMC10661996 DOI: 10.1016/j.obpill.2022.100053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/06/2022] [Accepted: 12/22/2022] [Indexed: 11/23/2023]
Abstract
Objective The number of children with cardiovascular risk factors is increasing steadily. However, limited data are available on the prevalence of overweight, obesity, and hyperglycemia among children in low-middle-income countries with multiethnic populations. Therefore, we assessed these factors in a school-based survey in Suriname, a low-middle-income country. Methods We invited pupils of 5th and 6th grade visiting the primary school to participate in this survey. We used a questionnaire and face to face interviews, and conducted measurements to collect data on biological factors (ethnicity, sex, length, weight, waist circumference, and fasting blood glucose levels), behavior (frequency of physical activity, breakfast, bedtime, screentime), consumption (fruit and vegetables, snack, dairy products) and social factors (parental education, living area). Results Overall, the percentage of children with overweight was 13.9%, obesity 13.3% and for elevated fasting blood glucose level (> 6 mmol/L) 4.5%. In the investigated group of individuals, obesity and overweight were associated with sex (girls showed a lower OR of 0.54 [95%CI: 0.39-0.75] for obesity), ethnicity (Javanese 2.1, 1.5-3.0 for overweight and 5.0, 3.1-8.2 for obesity, Maroon 2.2, 1.2-4.1 and Mixed ethnicity 1.7, 1.1-2.6, for obesity compared to Hindustani), behavior (Skip Breakfast: 1.4, 1.2-1.7, physical activity: 0.8, 0.7-0.9) and maternal education level (high 1.7, 1.0-2.7). Children with elevated fasting blood glucose levels showed an association with obesity (1.8, 1.2-2.7) and waist circumference (1.02, 1.01-1.03). Conclusion The results show that there is a high prevalence for overweight, obesity and elevated fasting blood glucose among children in Suriname. Furthermore, during childhood ethnicity is associated with obesity and overweight. We suggest that the modifiable risk factors such as BMI, WC, behavior, consumption are interesting for early intervention in children in a developing country.
Collapse
Affiliation(s)
- Jerry R. Toelsie
- Department of Physiology, Faculty of Medical Science, Anton de Kom University of Suriname, Paramaribo, Suriname
| | | | - Ingrid Krishnadath
- Department of Public Health, Faculty of Medical Science, Anton de Kom University of Suriname, Paramaribo, Suriname
| | - Robbert Bipat
- Department of Physiology, Faculty of Medical Science, Anton de Kom University of Suriname, Paramaribo, Suriname
| |
Collapse
|
35
|
Tribolet L, Brice AM, Fulford TS, Layton DS, Godfrey DI, Bean AGD, Stewart CR. Identification of a novel role for the immunomodulator ILRUN in the development of several T cell subsets in mice. Immunobiology 2023; 228:152380. [PMID: 37031606 DOI: 10.1016/j.imbio.2023.152380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/20/2023] [Accepted: 03/26/2023] [Indexed: 03/31/2023]
Abstract
Inflammation and lipid regulator with UBA-like and NBR1-like domains (ILRUN) is a protein-encoding gene associated with innate immune signaling, lipid metabolism and cancer. In the context of innate immunity, ILRUN inhibits IRF3-mediated transcription of antimicrobial and proinflammatory cytokines by inducing degradation of the transcriptional coactivators CBP and p300. There remains a paucity of information, however, regarding the innate immune roles of ILRUN beyond in vitro analyses. To address this, we utilize a knockout mouse model to investigate the effect of ILRUN on cytokine expression in splenocytes and on the development of immune cell populations in the spleen and thymus. We show elevated production of tumor necrosis factor and interleukin-6 cytokines in ILRUN-deficient splenocytes following stimulation with the innate immune ligands polyinosinic:polycytidylic acid or lipopolysaccharide. Differences were also observed in the populations of several T cell subsets, including regulatory, mucosal-associated invariant and natural killer. These data identify novel functions for ILRUN in the development of certain immune cell populations and support previous in vitro findings that ILRUN negatively regulates the synthesis of pathogen-stimulated cytokines. This establishes the ILRUN knockout mouse model as a valuable resource for further study of the functions of ILRUN in health and disease.
Collapse
Affiliation(s)
- Leon Tribolet
- CSIRO Health & Biosecurity, Australian Centre for Disease Preparedness, Geelong 3220, Victoria, Australia
| | - Aaron M Brice
- CSIRO Health & Biosecurity, Australian Centre for Disease Preparedness, Geelong 3220, Victoria, Australia
| | - Thomas S Fulford
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne 3000, Victoria, Australia
| | - Daniel S Layton
- CSIRO Health & Biosecurity, Australian Centre for Disease Preparedness, Geelong 3220, Victoria, Australia
| | - Dale I Godfrey
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne 3000, Victoria, Australia
| | - Andrew G D Bean
- CSIRO Health & Biosecurity, Australian Centre for Disease Preparedness, Geelong 3220, Victoria, Australia
| | - Cameron R Stewart
- CSIRO Health & Biosecurity, Australian Centre for Disease Preparedness, Geelong 3220, Victoria, Australia.
| |
Collapse
|
36
|
Tao L, Wang LG, Adeola AC, Zhang LC, Li LW, Li QL, Cen DJ, Yan C, Ma ZS, Wang LX, Xie HB, Zhang YP. Associations of autozygosity with economic important traits in a cross of Eurasian pigs. J Genet Genomics 2023; 50:216-220. [PMID: 36152906 DOI: 10.1016/j.jgg.2022.09.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 08/30/2022] [Accepted: 09/16/2022] [Indexed: 10/14/2022]
Affiliation(s)
- Lin Tao
- State Key Laboratory of Genetic Resources and Evolution Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Li-Gang Wang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Adeniyi C Adeola
- State Key Laboratory of Genetic Resources and Evolution Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Long-Chao Zhang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China
| | - Lian-Wei Li
- Computational Biology and Medical Ecology Lab, State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Qing-Long Li
- State Key Laboratory of Genetic Resources and Evolution Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; State Key Laboratory for Conservation and Utilization of Bio-resource in Yunnan, School of Life Science, Yunnan University, Kunming, Yunnan 650091, China
| | - Dao-Ji Cen
- State Key Laboratory of Genetic Resources and Evolution Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Chen Yan
- State Key Laboratory of Genetic Resources and Evolution Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China
| | - Zhan-Shan Ma
- Computational Biology and Medical Ecology Lab, State Key Laboratory of Genetic Resources and Evolution, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China
| | - Li-Xian Wang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China.
| | - Hai-Bing Xie
- State Key Laboratory of Genetic Resources and Evolution Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China.
| | - Ya-Ping Zhang
- State Key Laboratory of Genetic Resources and Evolution Yunnan Laboratory of Molecular Biology of Domestic Animals, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 650223, China; State Key Laboratory for Conservation and Utilization of Bio-resource in Yunnan, School of Life Science, Yunnan University, Kunming, Yunnan 650091, China; Kunming College of Life Science, University of Chinese Academy of Sciences, Kunming, Yunnan 650204, China.
| |
Collapse
|
37
|
Tsao CW, Aday AW, Almarzooq ZI, Anderson CAM, Arora P, Avery CL, Baker-Smith CM, Beaton AZ, Boehme AK, Buxton AE, Commodore-Mensah Y, Elkind MSV, Evenson KR, Eze-Nliam C, Fugar S, Generoso G, Heard DG, Hiremath S, Ho JE, Kalani R, Kazi DS, Ko D, Levine DA, Liu J, Ma J, Magnani JW, Michos ED, Mussolino ME, Navaneethan SD, Parikh NI, Poudel R, Rezk-Hanna M, Roth GA, Shah NS, St-Onge MP, Thacker EL, Virani SS, Voeks JH, Wang NY, Wong ND, Wong SS, Yaffe K, Martin SS. Heart Disease and Stroke Statistics-2023 Update: A Report From the American Heart Association. Circulation 2023; 147:e93-e621. [PMID: 36695182 DOI: 10.1161/cir.0000000000001123] [Citation(s) in RCA: 2289] [Impact Index Per Article: 1144.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND The American Heart Association, in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, diet, and weight) and health factors (cholesterol, blood pressure, and glucose control) that contribute to cardiovascular health. The Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, heart failure, valvular disease, venous disease, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The American Heart Association, through its Epidemiology and Prevention Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States to provide the most current information available in the annual Statistical Update with review of published literature through the year before writing. The 2023 Statistical Update is the product of a full year's worth of effort in 2022 by dedicated volunteer clinicians and scientists, committed government professionals, and American Heart Association staff members. The American Heart Association strives to further understand and help heal health problems inflicted by structural racism, a public health crisis that can significantly damage physical and mental health and perpetuate disparities in access to health care, education, income, housing, and several other factors vital to healthy lives. This year's edition includes additional COVID-19 (coronavirus disease 2019) publications, as well as data on the monitoring and benefits of cardiovascular health in the population, with an enhanced focus on health equity across several key domains. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|
38
|
Hong-Le T, Crouse WL, Keele GR, Holl K, Seshie O, Tschannen M, Craddock A, Das SK, Szalanczy AM, McDonald B, Grzybowski M, Klotz J, Sharma NK, Geurts AM, Key CCC, Hawkins G, Valdar W, Mott R, Solberg Woods LC. Genetic Mapping of Multiple Traits Identifies Novel Genes for Adiposity, Lipids, and Insulin Secretory Capacity in Outbred Rats. Diabetes 2023; 72:135-148. [PMID: 36219827 PMCID: PMC9797320 DOI: 10.2337/db22-0252] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 10/04/2022] [Indexed: 01/21/2023]
Abstract
Despite the successes of human genome-wide association studies, the causal genes underlying most metabolic traits remain unclear. We used outbred heterogeneous stock (HS) rats, coupled with expression data and mediation analysis, to identify quantitative trait loci (QTLs) and candidate gene mediators for adiposity, glucose tolerance, serum lipids, and other metabolic traits. Physiological traits were measured in 1,519 male HS rats, with liver and adipose transcriptomes measured in >410 rats. Genotypes were imputed from low-coverage whole-genome sequencing. Linear mixed models were used to detect physiological and expression QTLs (pQTLs and eQTLs, respectively), using both single nucleotide polymorphism (SNP)- and haplotype-based models for pQTL mapping. Genes with cis-eQTLs that overlapped pQTLs were assessed as causal candidates through mediation analysis. We identified 14 SNP-based pQTLs and 19 haplotype-based pQTLs, of which 10 were in common. Using mediation, we identified the following genes as candidate mediators of pQTLs: Grk5 for fat pad weight and serum triglyceride pQTLs on Chr1, Krtcap3 for fat pad weight and serum triglyceride pQTLs on Chr6, Ilrun for a fat pad weight pQTL on Chr20, and Rfx6 for a whole pancreatic insulin content pQTL on Chr20. Furthermore, we verified Grk5 and Ktrcap3 using gene knockdown/out models, thereby shedding light on novel regulators of obesity.
Collapse
Affiliation(s)
- Thu Hong-Le
- Genetics Institute, University College London, London, U.K
| | - Wesley L. Crouse
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | | - Katie Holl
- Medical College of Wisconsin, Milwaukee, WI
| | - Osborne Seshie
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
| | | | - Ann Craddock
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Swapan K. Das
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Alexandria M. Szalanczy
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Bailey McDonald
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
| | | | | | - Neeraj K. Sharma
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
| | | | - Chia-Chi Chuang Key
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Gregory Hawkins
- Department of Biochemistry, Wake Forest University School of Medicine, Winston-Salem, NC
| | - William Valdar
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Richard Mott
- Genetics Institute, University College London, London, U.K
| | - Leah C. Solberg Woods
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
| |
Collapse
|
39
|
Wojcik GL, Murphy J, Edelson JL, Gignoux CR, Ioannidis AG, Manning A, Rivas MA, Buyske S, Hendricks AE. Opportunities and challenges for the use of common controls in sequencing studies. Nat Rev Genet 2022; 23:665-679. [PMID: 35581355 PMCID: PMC9765323 DOI: 10.1038/s41576-022-00487-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2022] [Indexed: 01/02/2023]
Abstract
Genome-wide association studies using large-scale genome and exome sequencing data have become increasingly valuable in identifying associations between genetic variants and disease, transforming basic research and translational medicine. However, this progress has not been equally shared across all people and conditions, in part due to limited resources. Leveraging publicly available sequencing data as external common controls, rather than sequencing new controls for every study, can better allocate resources by augmenting control sample sizes or providing controls where none existed. However, common control studies must be carefully planned and executed as even small differences in sample ascertainment and processing can result in substantial bias. Here, we discuss challenges and opportunities for the robust use of common controls in high-throughput sequencing studies, including study design, quality control and statistical approaches. Thoughtful generation and use of large and valuable genetic sequencing data sets will enable investigation of a broader and more representative set of conditions, environments and genetic ancestries than otherwise possible.
Collapse
Affiliation(s)
- Genevieve L Wojcik
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Jessica Murphy
- Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, USA
- Mathematical and Statistical Sciences, University of Colorado Denver, Denver, CO, USA
| | - Jacob L Edelson
- Department of Biomedical Data Science, Stanford Medical School, Stanford, CA, USA
| | - Christopher R Gignoux
- Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, USA
- Human Medical Genetics and Genomics Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Alexander G Ioannidis
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA, USA
| | - Alisa Manning
- Metabolism Program, Broad Institute, Cambridge, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Manuel A Rivas
- Department of Biomedical Data Science, Stanford Medical School, Stanford, CA, USA
| | - Steven Buyske
- Department of Statistics, Rutgers University, Piscataway, NJ, USA
| | - Audrey E Hendricks
- Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO, USA.
- Mathematical and Statistical Sciences, University of Colorado Denver, Denver, CO, USA.
- Human Medical Genetics and Genomics Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
- Colorado Center for Personalized Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
40
|
Tan PY, Moore JB, Bai L, Tang G, Gong YY. In the context of the triple burden of malnutrition: A systematic review of gene-diet interactions and nutritional status. Crit Rev Food Sci Nutr 2022; 64:3235-3263. [PMID: 36222100 PMCID: PMC11000749 DOI: 10.1080/10408398.2022.2131727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Genetic background interacts with dietary components to modulate nutritional health status. This study aimed to review the evidence for gene-diet interactions in all forms of malnutrition. A comprehensive systematic literature search was conducted through April 2021 to identify observational and intervention studies reporting the effects of gene-diet interactions in over-nutrition, under-nutrition and micronutrient status. Risk of publication bias was assessed using the Quality Criteria Checklist and a tool specifically designed for gene-diet interaction research. 167 studies from 27 populations were included. The majority of studies investigated single nucleotide polymorphisms (SNPs) in overnutrition (n = 158). Diets rich in whole grains, vegetables, fruits and low in total and saturated fats, such as Mediterranean and DASH diets, showed promising effects for reducing obesity risk among individuals who had higher genetic risk scores for obesity, particularly the risk alleles carriers of FTO rs9939609, rs1121980 and rs1421085. Other SNPs in MC4R, PPARG and APOA5 genes were also commonly studied for interaction with diet on overnutrition though findings were inconclusive. Only limited data were found related to undernutrition (n = 1) and micronutrient status (n = 9). The findings on gene-diet interactions in this review highlight the importance of personalized nutrition, and more research on undernutrition and micronutrient status is warranted.
Collapse
Affiliation(s)
- Pui Yee Tan
- School of Food Science and Nutrition, Faculty of Environment, University of Leeds, Leeds, United Kingdom
| | - J. Bernadette Moore
- School of Food Science and Nutrition, Faculty of Environment, University of Leeds, Leeds, United Kingdom
| | - Ling Bai
- School of Food Science and Nutrition, Faculty of Environment, University of Leeds, Leeds, United Kingdom
- School of Psychology, University of East Anglia, Norwich, United Kingdom
| | - GuYuan Tang
- School of Food Science and Nutrition, Faculty of Environment, University of Leeds, Leeds, United Kingdom
| | - Yun Yun Gong
- School of Food Science and Nutrition, Faculty of Environment, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
41
|
Hu S, Zhang X, Stamatiou M, Hambly C, Huang Y, Ma J, Li Y, Speakman JR. Higher than predicted resting energy expenditure and lower physical activity in healthy underweight Chinese adults. Cell Metab 2022; 34:1413-1415. [PMID: 35839758 DOI: 10.1016/j.cmet.2022.05.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 03/03/2022] [Accepted: 05/27/2022] [Indexed: 11/03/2022]
Abstract
Contrary to popular opinion that lean individuals "eat what they want" and exercise more, Hu et al. study a cohort of healthy underweight volunteers and reveal them to have reduced physical activity relative to normal BMI controls and lower food intake. This cohort is also shown to have higher than expected resting energy expenditure, which is associated with elevations in thyroid hormones.
Collapse
Affiliation(s)
- Sumei Hu
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PRC; State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, PRC; Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Engineering and Technology Research Center of Food Additives, National Soybean Processing Industry Technology Innovation Center, Beijing Technology and Business University, Beijing 100048, PRC
| | - Xueying Zhang
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PRC; State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, PRC; School of Biological Sciences, University of Aberdeen, Aberdeen, AB24 2TZ, Scotland, UK
| | - Marina Stamatiou
- School of Biological Sciences, University of Aberdeen, Aberdeen, AB24 2TZ, Scotland, UK
| | - Catherine Hambly
- School of Biological Sciences, University of Aberdeen, Aberdeen, AB24 2TZ, Scotland, UK
| | - Yumeng Huang
- Department of Neurology, Institute of Neurology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, PRC
| | - Jianfang Ma
- Department of Neurology, Institute of Neurology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, PRC
| | - Yiran Li
- College of Biology, China Agricultural University, Beijing, PRC
| | - John R Speakman
- Shenzhen Key Laboratory of Metabolic Health, Center for Energy Metabolism and Reproduction, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PRC; State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, PRC; School of Biological Sciences, University of Aberdeen, Aberdeen, AB24 2TZ, Scotland, UK; CAS Center for Excellence in Animal Evolution and Genetics (CCEAEG), Kunming, PRC.
| |
Collapse
|
42
|
Hinney A, Körner A, Fischer-Posovszky P. The promise of new anti-obesity therapies arising from knowledge of genetic obesity traits. Nat Rev Endocrinol 2022; 18:623-637. [PMID: 35902734 PMCID: PMC9330928 DOI: 10.1038/s41574-022-00716-0] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/17/2022] [Indexed: 02/07/2023]
Abstract
Obesity is a multifactorial and complex disease that often manifests in early childhood with a lifelong burden. Polygenic and monogenic obesity are driven by the interaction between genetic predisposition and environmental factors. Polygenic variants are frequent and confer small effect sizes. Rare monogenic obesity syndromes are caused by defined pathogenic variants in single genes with large effect sizes. Most of these genes are involved in the central nervous regulation of body weight; for example, genes of the leptin-melanocortin pathway. Clinically, patients with monogenic obesity present with impaired satiety, hyperphagia and pronounced food-seeking behaviour in early childhood, which leads to severe early-onset obesity. With the advent of novel pharmacological treatment options emerging for monogenic obesity syndromes that target the central melanocortin pathway, genetic testing is recommended for patients with rapid weight gain in infancy and additional clinical suggestive features. Likewise, patients with obesity associated with hypothalamic damage or other forms of syndromic obesity involving energy regulatory circuits could benefit from these novel pharmacological treatment options. Early identification of patients affected by syndromic obesity will lead to appropriate treatment, thereby preventing the development of obesity sequelae, avoiding failure of conservative treatment approaches and alleviating stigmatization of patients and their families.
Collapse
Affiliation(s)
- Anke Hinney
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy and University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
- Center for Translational Neuro- and Behavioral Sciences, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| | - Antje Körner
- Leipzig University, Medical Faculty, Hospital for Children and Adolescents, Centre of Paediatric Research (CPL), Leipzig, Germany
- LIFE Child, Leipzig Research Centre for Civilization Diseases, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | | |
Collapse
|
43
|
Eslam M, El-Serag HB, Francque S, Sarin SK, Wei L, Bugianesi E, George J. Metabolic (dysfunction)-associated fatty liver disease in individuals of normal weight. Nat Rev Gastroenterol Hepatol 2022; 19:638-651. [PMID: 35710982 DOI: 10.1038/s41575-022-00635-5] [Citation(s) in RCA: 134] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/13/2022] [Indexed: 12/12/2022]
Abstract
Metabolic (dysfunction)-associated fatty liver disease (MAFLD) affects up to a third of the global population; its burden has grown in parallel with rising rates of type 2 diabetes mellitus and obesity. MAFLD increases the risk of end-stage liver disease, hepatocellular carcinoma, death and liver transplantation and has extrahepatic consequences, including cardiometabolic disease and cancers. Although typically associated with obesity, there is accumulating evidence that not all people with overweight or obesity develop fatty liver disease. On the other hand, a considerable proportion of patients with MAFLD are of normal weight, indicating the importance of metabolic health in the pathogenesis of the disease regardless of body mass index. The clinical profile, natural history and pathophysiology of patients with so-called lean MAFLD are not well characterized. In this Review, we provide epidemiological data on this group of patients and consider overall metabolic health and metabolic adaptation as a framework to best explain the pathogenesis of MAFLD and its heterogeneity in individuals of normal weight and in those who are above normal weight. This framework provides a conceptual schema for interrogating the MAFLD phenotype in individuals of normal weight that can translate to novel approaches for diagnosis and patient care.
Collapse
Affiliation(s)
- Mohammed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia.
| | - Hashem B El-Serag
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Sven Francque
- Department of Gastroenterology and Hepatology, Antwerp University Hospital, Antwerp, Belgium
- Laboratory of Experimental Medicine and Paediatrics (LEMP), Faculty of Medicine and Health Sciences, University of Antwerp, Antwerp, Belgium
| | - Shiv K Sarin
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Lai Wei
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Elisabetta Bugianesi
- Department of Medical Sciences, Division of Gastroenterology and Hepatology, A.O. Città della Salute e della Scienza di Torino, University of Turin, Turin, Italy
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
44
|
Guilherme JPLF, Semenova EA, Larin AK, Yusupov RA, Generozov EV, Ahmetov II. Genomic Predictors of Brisk Walking Are Associated with Elite Sprinter Status. Genes (Basel) 2022; 13:genes13101710. [PMID: 36292594 PMCID: PMC9602420 DOI: 10.3390/genes13101710] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 11/16/2022] Open
Abstract
Brisk walkers are physically more active, taller, have reduced body fat and greater physical fitness and muscle strength. The aim of our study was to determine whether genetic variants associated with increased walking pace were overrepresented in elite sprinters compared to controls. A total of 70 single-nucleotide polymorphisms (SNPs) previously identified in a genome-wide association study (GWAS) of self-reported walking pace in 450,967 European individuals were explored in relation to sprinter status. Genotyping of 137 Russian elite sprinters and 126 controls was performed using microarray technology. Favorable (i.e., high-speed-walking) alleles of 15 SNPs (FHL2 rs55680124 C, SLC39A8 rs13107325 C, E2F3 rs4134943 T, ZNF568 rs1667369 A, GDF5 rs143384 G, PPARG rs2920503 T, AUTS2 rs10452738 A, IGSF3 rs699785 A, CCT3 rs11548200 T, CRTAC1 rs2439823 A, ADAM15 rs11264302 G, C6orf106 rs205262 A, AKAP6 rs12883788 C, CRTC1 rs11881338 A, NRXN3 rs8011870 G) were identified as having positive associations with sprinter status (p < 0.05), of which IGSF3 rs699785 survived correction for multiple testing (p = 0.00004) and was linked (p = 0.042) with increased proportions of fast-twitch muscle fibers of m. vastus lateralis in physically active men (n = 67). Polygenic analysis revealed that individuals with ≥18 favorable alleles of the 15 SNPs have an increased odds ratio of being an elite sprinter when compared to those with ≤17 alleles (OR: 7.89; p < 0.0001). Using UK Biobank data, we also established the association of 14 favorable alleles with low BMI and fat percentage, 8 alleles with increased handgrip strength, and 7 alleles with increased height and fat-free mass. In conclusion, we have identified 15 new genetic markers associated with sprinter status.
Collapse
Affiliation(s)
- João Paulo L. F. Guilherme
- Laboratory of Applied Nutrition and Metabolism, School of Physical Education and Sport, University of São Paulo, São Paulo 05508-030, Brazil
- Laboratory of Biochemistry and Molecular Biology of Exercise, School of Physical Education and Sport, University of São Paulo, São Paulo 05508-030, Brazil
- Correspondence: (J.P.L.F.G.); (I.I.A.)
| | - Ekaterina A. Semenova
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
- Research Institute of Physical Culture and Sport, Volga Region State University of Physical Culture, Sport and Tourism, 420138 Kazan, Russia
| | - Andrey K. Larin
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
| | - Rinat A. Yusupov
- Department of Physical Culture and Sport, Kazan National Research Technical University Named after A.N. Tupolev-KAI, 420111 Kazan, Russia
| | - Edward V. Generozov
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, 119435 Moscow, Russia
| | - Ildus I. Ahmetov
- Department of Physical Education, Plekhanov Russian University of Economics, 115093 Moscow, Russia
- Laboratory of Molecular Genetics, Central Research Laboratory, Kazan State Medical University, 420012 Kazan, Russia
- Sports Genetics Laboratory, St. Petersburg Research Institute of Physical Culture, 191040 St. Petersburg, Russia
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool L3 5AF, UK
- Correspondence: (J.P.L.F.G.); (I.I.A.)
| |
Collapse
|
45
|
Mahoney P, McFarlane G, Loch C, White S, Floyd B, Dunn EC, Pitfield R, Nava A, Guatelli-Steinberg D. Dental biorhythm is associated with adolescent weight gain. COMMUNICATIONS MEDICINE 2022; 2:99. [PMID: 36016726 PMCID: PMC9395425 DOI: 10.1038/s43856-022-00164-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 07/26/2022] [Indexed: 12/02/2022] Open
Abstract
Background Evidence of a long-period biological rhythm present in mammalian hard tissue relates to species average body mass. Studies have just begun to investigate the role of this biorhythm in human physiology. Methods The biorhythm is calculated from naturally exfoliated primary molars for 61 adolescents. We determine if the timing relates to longitudinal measures of their weight, height, lower leg length and body mass collected over 14 months between September 2019 to October 2020. We use univariate and multivariate statistical analyses to isolate and identify relationships with the biorhythm. Results Participants with a faster biorhythm typically weigh less each month and gain significantly less weight and mass over 14-months, relative to those with a slower biorhythm. The biorhythm relates to sex differences in weight gain. Conclusions We identify a previously unknown factor that associates with the rapid change in body size that accompanies human adolescence. Our findings provide a basis from which to explore novel relationships between the biorhythm and weight-related health risks.
Collapse
Affiliation(s)
- Patrick Mahoney
- School of Anthropology and Conservation, University of Kent, Canterbury, UK
| | - Gina McFarlane
- School of Anthropology and Conservation, University of Kent, Canterbury, UK
| | - Carolina Loch
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Sophie White
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin, New Zealand
| | - Bruce Floyd
- School of Social Sciences, University of Auckland, Auckland, New Zealand
| | - Erin C. Dunn
- Department of Psychiatry, Harvard Medical School and the Massachusetts General Hospital, Boston, MA USA
| | - Rosie Pitfield
- School of Anthropology and Conservation, University of Kent, Canterbury, UK
| | - Alessia Nava
- School of Anthropology and Conservation, University of Kent, Canterbury, UK
| | - Debbie Guatelli-Steinberg
- School of Anthropology and Conservation, University of Kent, Canterbury, UK
- Department of Anthropology, The Ohio State University, Columbus, OH USA
| |
Collapse
|
46
|
Azzolini F, Berentsen GD, Skaug HJ, Hjelmborg JVB, Kaprio JA. The heritability of BMI varies across the range of BMI-a heritability curve analysis in a twin cohort. Int J Obes (Lond) 2022; 46:1786-1791. [PMID: 35817850 DOI: 10.1038/s41366-022-01172-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 06/09/2022] [Accepted: 06/14/2022] [Indexed: 12/18/2022]
Abstract
BACKGROUND The heritability of traits such as body mass index (BMI), a measure of obesity, is generally estimated using family and twin studies, and increasingly by molecular genetic approaches. These studies generally assume that genetic effects are uniform across all trait values, yet there is emerging evidence that this may not always be the case. METHOD/SUBJECTS This paper analyzes twin data using a recently developed measure of heritability called the heritability curve. Under the assumption that trait values in twin pairs are governed by a flexible Gaussian mixture distribution, heritability curves may vary across trait values. The data consist of repeated measures of BMI on 1506 monozygotic (MZ) and 2843 like-sexed dizygotic (DZ) adult twin pairs, gathered from multiple surveys in older Finnish Twin Cohorts. RESULTS The heritability curve and BMI value-specific MZ and DZ pairwise correlations were estimated, and these varied across the range of BMI. MZ correlations were highest at BMI values from 21 to 24, with a stronger decrease for women than for men at higher values. Models with additive and dominance effects fit best at low and high BMI values, while models with additive genetic and common environmental effects fit best in the normal range of BMI. CONCLUSIONS We demonstrate that twin and molecular genetic studies need to consider how genetic effects vary across trait values. Such variation may reconcile findings of traits with high heritability and major differences in mean values between countries or over time.
Collapse
Affiliation(s)
| | - Geir D Berentsen
- Department of Business and Management Science, NHH Norwegian School of Economics, Bergen, Norway
| | - Hans J Skaug
- Department of Mathematics, University of Bergen, Bergen, Norway
| | - Jacob V B Hjelmborg
- Department of Public Health, University of Southern Denmark, Odense, Denmark
| | - Jaakko A Kaprio
- Institute for Molecular Medicine Finland, University of Helsinki, Helsinki, Finland
| |
Collapse
|
47
|
Ahmed M, Kaur N, Cheng Q, Shanabrough M, Tretiakov EO, Harkany T, Horvath TL, Schlessinger J. A hypothalamic pathway for Augmentor α-controlled body weight regulation. Proc Natl Acad Sci U S A 2022; 119:e2200476119. [PMID: 35412887 PMCID: PMC9169862 DOI: 10.1073/pnas.2200476119] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 03/15/2022] [Indexed: 11/29/2022] Open
Abstract
Augmentor α and β (Augα and Augβ) are newly discovered ligands of the receptor tyrosine kinases Alk and Ltk. Augα functions as a dimeric ligand that binds with high affinity and specificity to Alk and Ltk. However, a monomeric Augα fragment and monomeric Augβ also bind to Alk and potently stimulate cellular responses. While previous studies demonstrated that oncogenic Alk mutants function as important drivers of a variety of human cancers, the physiological roles of Augα and Augβ are poorly understood. Here, we investigate the physiological roles of Augα and Augβ by exploring mice deficient in each or both Aug ligands. Analysis of mutant mice showed that both Augα single knockout and double knockout of Augα and Augβ exhibit a similar thinness phenotype and resistance to diet-induced obesity. In the Augα-knockout mice, the leanness phenotype is coupled to increased physical activity. By contrast, Augβ-knockout mice showed similar weight curves as the littermate controls. Experiments are presented demonstrating that Augα is robustly expressed and metabolically regulated in agouti-related peptide (AgRP) neurons, cells that control whole-body energy homeostasis in part via their projections to the paraventricular nucleus (PVN). Moreover, both Alk and melanocortin receptor-4 are expressed in discrete neuronal populations in the PVN and are regulated by projections containing Augα and AgRP, respectively, demonstrating that two distinct mechanisms that regulate pigmentation operate in the hypothalamus to control body weight. These experiments show that Alk-driven cancers were co-opted from a neuronal pathway in control of body weight, offering therapeutic opportunities for metabolic diseases and cancer.
Collapse
Affiliation(s)
- Mansoor Ahmed
- Department of Pharmacology, Yale School of Medicine, New Haven, CT 06520
| | - Navjot Kaur
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06520
| | - Qianni Cheng
- Department of Pharmacology, Yale School of Medicine, New Haven, CT 06520
| | - Marya Shanabrough
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520
| | - Evgenii O. Tretiakov
- Department of Molecular Neurosciences, Medical University of Vienna, 1010 Vienna, Austria
| | - Tibor Harkany
- Department of Molecular Neurosciences, Medical University of Vienna, 1010 Vienna, Austria
- Department of Neuroscience, Karolinska Institutet, 17177 Solna, Sweden
| | - Tamas L. Horvath
- Department of Neuroscience, Yale School of Medicine, New Haven, CT 06520
- Department of Comparative Medicine, Yale School of Medicine, New Haven, CT 06520
| | | |
Collapse
|
48
|
Fernández-Rhodes L, Graff M, Buchanan VL, Justice AE, Highland HM, Guo X, Zhu W, Chen HH, Young KL, Adhikari K, Palmer ND, Below JE, Bradfield J, Pereira AC, Glover L, Kim D, Lilly AG, Shrestha P, Thomas AG, Zhang X, Chen M, Chiang CW, Pulit S, Horimoto A, Krieger JE, Guindo-Martínez M, Preuss M, Schumann C, Smit RA, Torres-Mejía G, Acuña-Alonzo V, Bedoya G, Bortolini MC, Canizales-Quinteros S, Gallo C, González-José R, Poletti G, Rothhammer F, Hakonarson H, Igo R, Adler SG, Iyengar SK, Nicholas SB, Gogarten SM, Isasi CR, Papnicolaou G, Stilp AM, Qi Q, Kho M, Smith JA, Langefeld CD, Wagenknecht L, Mckean-Cowdin R, Gao XR, Nousome D, Conti DV, Feng Y, Allison MA, Arzumanyan Z, Buchanan TA, Ida Chen YD, Genter PM, Goodarzi MO, Hai Y, Hsueh W, Ipp E, Kandeel FR, Lam K, Li X, Nadler JL, Raffel LJ, Roll K, Sandow K, Tan J, Taylor KD, Xiang AH, Yao J, Audirac-Chalifour A, de Jesus Peralta Romero J, Hartwig F, Horta B, Blangero J, Curran JE, Duggirala R, Lehman DE, Puppala S, Fejerman L, John EM, Aguilar-Salinas C, Burtt NP, Florez JC, García-Ortíz H, González-Villalpando C, Mercader J, Orozco L, Tusié-Luna T, Blanco E, Gahagan S, Cox NJ, Hanis C, et alFernández-Rhodes L, Graff M, Buchanan VL, Justice AE, Highland HM, Guo X, Zhu W, Chen HH, Young KL, Adhikari K, Palmer ND, Below JE, Bradfield J, Pereira AC, Glover L, Kim D, Lilly AG, Shrestha P, Thomas AG, Zhang X, Chen M, Chiang CW, Pulit S, Horimoto A, Krieger JE, Guindo-Martínez M, Preuss M, Schumann C, Smit RA, Torres-Mejía G, Acuña-Alonzo V, Bedoya G, Bortolini MC, Canizales-Quinteros S, Gallo C, González-José R, Poletti G, Rothhammer F, Hakonarson H, Igo R, Adler SG, Iyengar SK, Nicholas SB, Gogarten SM, Isasi CR, Papnicolaou G, Stilp AM, Qi Q, Kho M, Smith JA, Langefeld CD, Wagenknecht L, Mckean-Cowdin R, Gao XR, Nousome D, Conti DV, Feng Y, Allison MA, Arzumanyan Z, Buchanan TA, Ida Chen YD, Genter PM, Goodarzi MO, Hai Y, Hsueh W, Ipp E, Kandeel FR, Lam K, Li X, Nadler JL, Raffel LJ, Roll K, Sandow K, Tan J, Taylor KD, Xiang AH, Yao J, Audirac-Chalifour A, de Jesus Peralta Romero J, Hartwig F, Horta B, Blangero J, Curran JE, Duggirala R, Lehman DE, Puppala S, Fejerman L, John EM, Aguilar-Salinas C, Burtt NP, Florez JC, García-Ortíz H, González-Villalpando C, Mercader J, Orozco L, Tusié-Luna T, Blanco E, Gahagan S, Cox NJ, Hanis C, Butte NF, Cole SA, Comuzzie AG, Voruganti VS, Rohde R, Wang Y, Sofer T, Ziv E, Grant SF, Ruiz-Linares A, Rotter JI, Haiman CA, Parra EJ, Cruz M, Loos RJ, North KE. Ancestral diversity improves discovery and fine-mapping of genetic loci for anthropometric traits-The Hispanic/Latino Anthropometry Consortium. HGG ADVANCES 2022; 3:100099. [PMID: 35399580 PMCID: PMC8990175 DOI: 10.1016/j.xhgg.2022.100099] [Show More Authors] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 03/06/2022] [Indexed: 02/05/2023] Open
Abstract
Hispanic/Latinos have been underrepresented in genome-wide association studies (GWAS) for anthropometric traits despite their notable anthropometric variability, ancestry proportions, and high burden of growth stunting and overweight/obesity. To address this knowledge gap, we analyzed densely imputed genetic data in a sample of Hispanic/Latino adults to identify and fine-map genetic variants associated with body mass index (BMI), height, and BMI-adjusted waist-to-hip ratio (WHRadjBMI). We conducted a GWAS of 18 studies/consortia as part of the Hispanic/Latino Anthropometry (HISLA) Consortium (stage 1, n = 59,771) and generalized our findings in 9 additional studies (stage 2, n = 10,538). We conducted a trans-ancestral GWAS with summary statistics from HISLA stage 1 and existing consortia of European and African ancestries. In our HISLA stage 1 + 2 analyses, we discovered one BMI locus, as well as two BMI signals and another height signal each within established anthropometric loci. In our trans-ancestral meta-analysis, we discovered three BMI loci, one height locus, and one WHRadjBMI locus. We also identified 3 secondary signals for BMI, 28 for height, and 2 for WHRadjBMI in established loci. We show that 336 known BMI, 1,177 known height, and 143 known WHRadjBMI (combined) SNPs demonstrated suggestive transferability (nominal significance and effect estimate directional consistency) in Hispanic/Latino adults. Of these, 36 BMI, 124 height, and 11 WHRadjBMI SNPs were significant after trait-specific Bonferroni correction. Trans-ancestral meta-analysis of the three ancestries showed a small-to-moderate impact of uncorrected population stratification on the resulting effect size estimates. Our findings demonstrate that future studies may also benefit from leveraging diverse ancestries and differences in linkage disequilibrium patterns to discover novel loci and additional signals with less residual population stratification.
Collapse
Affiliation(s)
- Lindsay Fernández-Rhodes
- Department of Biobehavioral Health, Pennsylvania State University, 219 Biobehavioral Health Building, University Park, PA 16802, USA
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mariaelisa Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Victoria L. Buchanan
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Anne E. Justice
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Biomedical and Translational Informatics, Geisinger Health System, Danville, PA 17822, USA
| | - Heather M. Highland
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502 USA
| | - Wanying Zhu
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Hung-Hsin Chen
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Kristin L. Young
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Kaustubh Adhikari
- School of Mathematics and Statistics, Faculty of Science, Technology, Engineering and Mathematics, The Open University, MK7 6AA Milton Keynes, UK
| | - Nicholette D. Palmer
- Department of Biochemistry, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Jennifer E. Below
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jonathan Bradfield
- Center for Applied Genomics, Division of Human Genetics, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Alexandre C. Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo, São Paulo 05508-220, Brazil
| | - LáShauntá Glover
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Daeeun Kim
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Adam G. Lilly
- Department of Sociology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Poojan Shrestha
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Division of Pediatric and Public Health, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Alvin G. Thomas
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xinruo Zhang
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Minhui Chen
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Charleston W.K. Chiang
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Quantitative and Computational Biology, University of Southern California, Los Angeles, CA 90007, USA
| | - Sara Pulit
- Vertex Pharmaceuticals, W2 6BD Oxford, UK
| | - Andrea Horimoto
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo, São Paulo 05508-220, Brazil
| | - Jose E. Krieger
- Laboratory of Genetics and Molecular Cardiology, Heart Institute, University of São Paulo, São Paulo 05508-220, Brazil
| | - Marta Guindo-Martínez
- The Charles Bronfman Institutes for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- The Novo Nordisk Center for Basic Metabolic Research, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Michael Preuss
- The Charles Bronfman Institutes for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Claudia Schumann
- Hasso Plattner Institute, University of Potsdam, Digital Health Center, 14482 Potsdam, Germany
| | - Roelof A.J. Smit
- The Charles Bronfman Institutes for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Gabriela Torres-Mejía
- Department of Research in Cardiovascular Diseases, Diabetes Mellitus, and Cancer, Population Health Research Center, National Institute of Public Health, Cuernavaca, Morelos 62100, Mexico
| | | | - Gabriel Bedoya
- Molecular Genetics Investigation Group, University of Antioquia, Medellín 1226, Colombia
| | - Maria-Cátira Bortolini
- Department of Genetics, Federal University of Rio Grande do Sul, Porto Alegre 90040-060, Brazil
| | - Samuel Canizales-Quinteros
- Population Genomics Applied to Health Unit, The National Institute of Genomic Medicine and the Faculty of Chemistry at the National Autonomous University of Mexico, Mexico City 04510, Mexico
| | - Carla Gallo
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima 15102, Peru
| | - Rolando González-José
- Patagonian Institute of the Social and Human Sciences, Patagonian National Center, Puerto Madryn U9120, Argentina
| | - Giovanni Poletti
- Laboratorios de Investigación y Desarrollo, Facultad de Ciencias y Filosofía, Universidad Peruana Cayetano Heredia, Lima 15102, Peru
| | | | - Hakon Hakonarson
- Center for Applied Genomics, Division of Human Genetics, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Robert Igo
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Sharon G. Adler
- Division of Nephrology and Hypertension, Harbor-University of California Los Angeles Medical Center, Torrance, CA 90502, USA
| | - Sudha K. Iyengar
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Susanne B. Nicholas
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
| | | | - Carmen R. Isasi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | | - Adrienne M. Stilp
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Minjung Kho
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Jennifer A. Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI 48109, USA
| | - Carl D. Langefeld
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Lynne Wagenknecht
- Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Roberta Mckean-Cowdin
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90032, USA
| | - Xiaoyi Raymond Gao
- Department of Ophthalmology and Visual Sciences, Department of Biomedical Informatics, Division of Human Genetics, The Ohio State University, Columbus, OH 43210, USA
| | - Darryl Nousome
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90032, USA
| | - David V. Conti
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Ye Feng
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90032, USA
| | - Matthew A. Allison
- Department of Family Medicine, University of California, San Diego, CA 92161, USA
| | - Zorayr Arzumanyan
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502 USA
| | - Thomas A. Buchanan
- Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502 USA
| | - Pauline M. Genter
- Department of Medicine, Division of Endocrinology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Mark O. Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Yang Hai
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502 USA
| | - Willa Hsueh
- Department of Internal Medicine, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Eli Ipp
- Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, CA 90095, USA
- Department of Medicine, Division of Endocrinology, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502, USA
| | - Fouad R. Kandeel
- Department of Translational Research & Cellular Therapeutics, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Kelvin Lam
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502 USA
| | - Xiaohui Li
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502 USA
| | - Jerry L. Nadler
- Department of Pharmacology at New York Medical College School of Medicine, Valhalla, NY 10595, USA
| | - Leslie J. Raffel
- Division of Genetic and Genomic Medicine, Department of Pediatrics, University of California, Irvine, CA 92697, USA
| | - Kathryn Roll
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502 USA
| | - Kevin Sandow
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502 USA
| | - Jingyi Tan
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502 USA
| | - Kent D. Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502 USA
| | - Anny H. Xiang
- Research and Evaluation Branch, Kaiser Permanente of Southern California, Pasadena, CA 91101, USA
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502 USA
| | - Astride Audirac-Chalifour
- Medical Research Unit in Biochemistry, Specialty Hospital, National Medical Center of the Twenty-First Century, Mexican Institute of Social Security, Mexico City 06725, Mexico
| | - Jose de Jesus Peralta Romero
- Medical Research Unit in Biochemistry, Specialty Hospital, National Medical Center of the Twenty-First Century, Mexican Institute of Social Security, Mexico City 06725, Mexico
| | - Fernando Hartwig
- Postgraduate Program in Epidemiology, Federal University of Pelotas, Pelotas 96010-610, Brazil
| | - Bernando Horta
- Postgraduate Program in Epidemiology, Federal University of Pelotas, Pelotas 96010-610, Brazil
| | - John Blangero
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville and Edinburg, TX 78520 and 78539, 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 and Edinburg, TX 78520 and 78539, USA
| | - Ravindranath Duggirala
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, School of Medicine, University of Texas Rio Grande Valley, Brownsville and Edinburg, TX 78520 and 78539, USA
| | - Donna E. Lehman
- Department of Medicine, School of Medicine, University of Texas Health San Antonio, San Antonio, TX 78229, USA
| | - Sobha Puppala
- Department of Internal Medicine, Section of Molecular Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27109, USA
| | - Laura Fejerman
- Department of Public Health Sciences, School of Medicine, and the Comprehensive Cancer Center, University of California Davis, Davis, CA 95616, USA
| | - Esther M. John
- Departments of Epidemiology & Population Health and Medicine-Oncology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Carlos Aguilar-Salinas
- Division of Nutrition, Salvador Zubirán National Institute of Health Sciences and Nutrition, Mexico City 14080, Mexico
| | - Noël P. Burtt
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
| | - Jose C. Florez
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Humberto García-Ortíz
- Laboratory of Immunogenomics and Metabolic Diseases, National Institute of Genomic Medicine, Mexico City 14610, Mexico
| | - Clicerio González-Villalpando
- Center for Diabetes Studies, Research Unit for Diabetes and Cardiovascular Risk, Center for Population Health Studies, National Institute of Public Health, Mexico City 14080, Mexico
| | - Josep Mercader
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of the Massachusetts Institute of Technology and Harvard, Cambridge, MA 02142, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Lorena Orozco
- Laboratory of Immunogenomics and Metabolic Diseases, National Institute of Genomic Medicine, Mexico City 14610, Mexico
| | - Teresa Tusié-Luna
- Molecular Biology and Medical Genomics Unity, Institute of Biomedical Research, The National Autonomous University of Mexico and the Salvador Zubirán National Institute of Health Sciences and Nutrition, Mexico City 14080, Mexico
| | - Estela Blanco
- Center for Community Health, Division of Academic General Pediatrics, University of California at San Diego, San Diego, CA 92093, USA
| | - Sheila Gahagan
- Center for Community Health, Division of Academic General Pediatrics, University of California at San Diego, San Diego, CA 92093, USA
| | - Nancy J. Cox
- Vanderbilt Genetics Institute, Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Craig Hanis
- University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Nancy F. Butte
- United States Department of Agriculture, Agricultural Research Service, The Children’s Nutrition Research Center, and the Department Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Shelley A. Cole
- Population Health Program, Texas Biomedical Research Institute, San Antonio, TX 78227, USA
| | | | - V. Saroja Voruganti
- Department of Nutrition and Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, NC 28081, USA
| | - Rebecca Rohde
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Yujie Wang
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Tamar Sofer
- Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
- Division of Sleep and Circadian Disorders, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Elad Ziv
- Division of General Internal Medicine, Department of Medicine, Helen Diller Family Comprehensive Cancer Center, Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94115, USA
| | - Struan F.A. Grant
- Center for Applied Genomics, Division of Human Genetics, Department of Pediatrics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Andres Ruiz-Linares
- Ministry of Education Key Laboratory of Contemporary Anthropology and Collaborative Innovation Center of Genetics and Development, School of Life Sciences and Human Phenome Institute, Fudan University, Shanghai 200438, China
- Department of Genetics, Evolution and Environment, and Genetics Institute of the University College London, London WC1E 6BT, UK
- Laboratory of Biocultural Anthropology, Law, Ethics, and Health, Aix-Marseille University, Marseille 13385, France
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA 90502 USA
| | - Christopher A. Haiman
- Center for Genetic Epidemiology, Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Esteban J. Parra
- Department of Anthropology, University of Toronto- Mississauga, Mississauga, ON L5L 1C6, Canada
| | - Miguel Cruz
- Medical Research Unit in Biochemistry, Specialty Hospital, National Medical Center of the Twenty-First Century, Mexican Institute of Social Security, Mexico City 06725, Mexico
| | - Ruth J.F. Loos
- The Charles Bronfman Institutes for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, 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 27599, USA
- Carolina Center for Genome Sciences, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| |
Collapse
|
49
|
Saeed S, Janjua QM, Haseeb A, Khanam R, Durand E, Vaillant E, Ning L, Badreddine A, Berberian L, Boissel M, Amanzougarene S, Canouil M, Derhourhi M, Bonnefond A, Arslan M, Froguel P. Rare Variant Analysis of Obesity-Associated Genes in Young Adults With Severe Obesity From a Consanguineous Population of Pakistan. Diabetes 2022; 71:694-705. [PMID: 35061034 DOI: 10.2337/db21-0373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 01/18/2022] [Indexed: 11/13/2022]
Abstract
Recent advances in genetic analysis have significantly helped in progressively attenuating the heritability gap of obesity and have brought into focus monogenic variants that disrupt the melanocortin signaling. In a previous study, next-generation sequencing revealed a monogenic etiology in ∼50% of the children with severe obesity from a consanguineous population in Pakistan. Here we assess rare variants in obesity-causing genes in young adults with severe obesity from the same region. Genomic DNA from 126 randomly selected young adult obese subjects (BMI 37.2 ± 0.3 kg/m2; age 18.4 ± 0.3 years) was screened by conventional or augmented whole-exome analysis for point mutations and copy number variants (CNVs). Leptin, insulin, and cortisol levels were measured by ELISA. We identified 13 subjects carrying 13 different pathogenic or likely pathogenic variants in LEPR, PCSK1, MC4R, NTRK2, POMC, SH2B1, and SIM1. We also identified for the first time in the human, two homozygous stop-gain mutations in ASNSD1 and IFI16 genes. Inactivation of these genes in mouse models has been shown to result in obesity. Additionally, we describe nine homozygous mutations (seven missense, one stop-gain, and one stop-loss) and four copy-loss CNVs in genes or genomic regions previously linked to obesity-associated traits by genome-wide association studies. Unexpectedly, in contrast to obese children, pathogenic mutations in LEP and LEPR were either absent or rare in this cohort of young adults. High morbidity and mortality risks and social disadvantage of children with LEP or LEPR deficiency may in part explain this difference between the two cohorts.
Collapse
Affiliation(s)
- Sadia Saeed
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, U.K
- Inserm UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille, France
- Lille University Hospital, University of Lille, Lille, France
| | - Qasim M Janjua
- Department of Physiology and Biophysics, National University of Science and Technology, Sohar, Oman
| | - Attiya Haseeb
- School of Life Sciences, Forman Christian College, Lahore, Pakistan
| | - Roohia Khanam
- School of Life Sciences, Forman Christian College, Lahore, Pakistan
| | - Emmanuelle Durand
- Inserm UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille, France
- Lille University Hospital, University of Lille, Lille, France
| | - Emmanuel Vaillant
- Inserm UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille, France
- Lille University Hospital, University of Lille, Lille, France
| | - Lijiao Ning
- Inserm UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille, France
- Lille University Hospital, University of Lille, Lille, France
| | - Alaa Badreddine
- Inserm UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille, France
- Lille University Hospital, University of Lille, Lille, France
| | - Lionel Berberian
- Inserm UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille, France
- Lille University Hospital, University of Lille, Lille, France
| | - Mathilde Boissel
- Inserm UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille, France
- Lille University Hospital, University of Lille, Lille, France
| | - Souhila Amanzougarene
- Inserm UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille, France
- Lille University Hospital, University of Lille, Lille, France
| | - Mickaël Canouil
- Inserm UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille, France
- Lille University Hospital, University of Lille, Lille, France
| | - Mehdi Derhourhi
- Inserm UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille, France
- Lille University Hospital, University of Lille, Lille, France
| | - Amélie Bonnefond
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, U.K
- Inserm UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille, France
- Lille University Hospital, University of Lille, Lille, France
| | - Muhammad Arslan
- School of Life Sciences, Forman Christian College, Lahore, Pakistan
| | - Philippe Froguel
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, U.K
- Inserm UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille, France
- Lille University Hospital, University of Lille, Lille, France
| |
Collapse
|
50
|
Tsao CW, Aday AW, Almarzooq ZI, Alonso A, Beaton AZ, Bittencourt MS, Boehme AK, Buxton AE, Carson AP, Commodore-Mensah Y, Elkind MSV, Evenson KR, Eze-Nliam C, Ferguson JF, Generoso G, Ho JE, Kalani R, Khan SS, Kissela BM, Knutson KL, Levine DA, Lewis TT, Liu J, Loop MS, Ma J, Mussolino ME, Navaneethan SD, Perak AM, Poudel R, Rezk-Hanna M, Roth GA, Schroeder EB, Shah SH, Thacker EL, VanWagner LB, Virani SS, Voecks JH, Wang NY, Yaffe K, Martin SS. Heart Disease and Stroke Statistics-2022 Update: A Report From the American Heart Association. Circulation 2022; 145:e153-e639. [PMID: 35078371 DOI: 10.1161/cir.0000000000001052] [Citation(s) in RCA: 3167] [Impact Index Per Article: 1055.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND The American Heart Association, in conjunction with the National Institutes of Health, annually reports the most up-to-date statistics related to heart disease, stroke, and cardiovascular risk factors, including core health behaviors (smoking, physical activity, diet, and weight) and health factors (cholesterol, blood pressure, and glucose control) that contribute to cardiovascular health. The Statistical Update presents the latest data on a range of major clinical heart and circulatory disease conditions (including stroke, congenital heart disease, rhythm disorders, subclinical atherosclerosis, coronary heart disease, heart failure, valvular disease, venous disease, and peripheral artery disease) and the associated outcomes (including quality of care, procedures, and economic costs). METHODS The American Heart Association, through its Statistics Committee, continuously monitors and evaluates sources of data on heart disease and stroke in the United States to provide the most current information available in the annual Statistical Update. The 2022 Statistical Update is the product of a full year's worth of effort by dedicated volunteer clinicians and scientists, committed government professionals, and American Heart Association staff members. This year's edition includes data on the monitoring and benefits of cardiovascular health in the population and an enhanced focus on social determinants of health, adverse pregnancy outcomes, vascular contributions to brain health, and the global burden of cardiovascular disease and healthy life expectancy. RESULTS Each of the chapters in the Statistical Update focuses on a different topic related to heart disease and stroke statistics. CONCLUSIONS The Statistical Update represents a critical resource for the lay public, policymakers, media professionals, clinicians, health care administrators, researchers, health advocates, and others seeking the best available data on these factors and conditions.
Collapse
|