1
|
Stuber GD, Schwitzgebel VM, Lüscher C. The neurobiology of overeating. Neuron 2025:S0896-6273(25)00182-5. [PMID: 40185087 DOI: 10.1016/j.neuron.2025.03.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/13/2024] [Accepted: 03/06/2025] [Indexed: 04/07/2025]
Abstract
Food intake serves to maintain energy homeostasis; however, overeating can result in obesity, which is associated with serious health complications. In this review, we explore the intricate relationship between overeating, obesity, and the underlying neurobiological mechanisms. We review the homeostatic and hedonic feeding systems, highlighting the role of the hypothalamus and reward systems in controlling food intake and energy balance. Dysregulation in both these systems leads to overeating, as seen in genetic syndromes and environmental models affecting appetite regulation when consuming highly palatable food. The concept of "food addiction" is examined, drawing parallels to drug addiction. We discuss the cellular substrate for addiction-related behavior and current pharmacological obesity treatments-in particular, GLP-1 receptor agonists-showcasing synaptic plasticity in the context of overeating and palatable food exposure. A comprehensive model integrating insights from addiction research is proposed to guide effective interventions for maladaptive feeding behaviors. Ultimately, unraveling the neurobiological basis of overeating holds promise for addressing the pressing public health issue of obesity.
Collapse
Affiliation(s)
- Garret D Stuber
- Center for the Neurobiology of Addiction, Pain, and Emotion, Department of Anesthesiology and Pain Medicine, Department of Pharmacology, University of Washington, Seattle, WA, USA
| | - Valerie M Schwitzgebel
- Pediatric Endocrinology and Diabetes Unit, Department of Pediatrics, Gynecology and Obstetrics, Geneva University Hospitals, 1211 Geneva, Switzerland; Institute of Genetics and Genomics (iGE3) in Geneva, University of Geneva, 1211 Geneva, Switzerland
| | - Christian Lüscher
- Institute of Genetics and Genomics (iGE3) in Geneva, University of Geneva, 1211 Geneva, Switzerland; Department of Basic Neurosciences, Medical Faculty, University of Geneva, 1211 Geneva, Switzerland; Clinic of Neurology, Department of Clinical Neurosciences, Geneva University Hospitals, 1211 Geneva, Switzerland; Synapsy Center for Mental Health Research, University of Geneva, 1211 Geneva, Switzerland.
| |
Collapse
|
2
|
Saeed S, Bonnefond A, Froguel P. Obesity: exploring its connection to brain function through genetic and genomic perspectives. Mol Psychiatry 2025; 30:651-658. [PMID: 39237720 PMCID: PMC11746128 DOI: 10.1038/s41380-024-02737-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/07/2024]
Abstract
Obesity represents an escalating global health burden with profound medical and economic impacts. The conventional perspective on obesity revolves around its classification as a "pure" metabolic disorder, marked by an imbalance between calorie consumption and energy expenditure. Present knowledge, however, recognizes the intricate interaction of rare or frequent genetic factors that favor the development of obesity, together with the emergence of neurodevelopmental and mental abnormalities, phenotypes that are modulated by environmental factors such as lifestyle. Thirty years of human genetic research has unveiled >20 genes, causing severe early-onset monogenic obesity and ~1000 loci associated with common polygenic obesity, most of those expressed in the brain, depicting obesity as a neurological and mental condition. Therefore, obesity's association with brain function should be better recognized. In this context, this review seeks to broaden the current perspective by elucidating the genetic determinants that contribute to both obesity and neurodevelopmental and mental dysfunctions. We conduct a detailed examination of recent genetic findings, correlating them with clinical and behavioral phenotypes associated with obesity. This includes how polygenic obesity, influenced by a myriad of genetic variants, impacts brain regions associated with addiction and reward, differentiating it from monogenic forms. The continuum between non-syndromic and syndromic monogenic obesity, with evidence from neurodevelopmental and cognitive assessments, is also addressed. Current therapeutic approaches that target these genetic mechanisms, yielding improved clinical outcomes and cognitive advantages, are discussed. To sum up, this review corroborates the genetic underpinnings of obesity, affirming its classification as a neurological disorder that may have broader implications for neurodevelopmental and mental conditions. It highlights the promising intersection of genetics, genomics, and neurobiology as a foundation for developing tailored medical approaches to treat obesity and its related neurological aspects.
Collapse
Affiliation(s)
- Sadia Saeed
- INSERM UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Lille, France
- University of Lille, Lille University Hospital, Lille, France
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Amélie Bonnefond
- INSERM UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Lille, France
- University of Lille, Lille University Hospital, Lille, France
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Philippe Froguel
- INSERM UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes (EGID), Lille, France.
- University of Lille, Lille University Hospital, Lille, France.
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK.
| |
Collapse
|
3
|
de Soysa AKH, Langaas M, Grill V, Martins C, Løvold Mostad I. Exploring associations between the FTO rs9939609 genotype and plasma concentrations of appetite-related hormones in adults with obesity. PLoS One 2025; 20:e0312815. [PMID: 39792913 PMCID: PMC11723609 DOI: 10.1371/journal.pone.0312815] [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: 10/14/2024] [Indexed: 01/12/2025] Open
Abstract
Associations between variants in the FTO locus and plasma concentrations of appetite related hormones are inconsistent, and might not work in a dose dependent fashion in people with obesity. Moreover, it is relevant to report meal related plasma concentrations of these hormones in persons with obesity given the growing interest in their pharmacological potential in obesity therapy. We find it clinically relevant to examine associations between the SNP rs9939609 genotypes and homeostatic appetite regulation in individuals with BMI ≥35 kg/m2. This study explored associations of the rs9939609 genotypes to plasma concentrations of acylated ghrelin, active glucagon-like peptide 1 (GLP-1), and total peptide YY (PYY), and moderating effects of fat mass (FM), in 96 adults (69% female) with BMI ≥35 kg/m2, using a cross sectional observation study designed to have 1/3 of participants each with genotypes TT, AT and AA, respectively. Participants were median (25th, 75th percentile) 42.5 (32, 50) years of age, weighed 120.9 (109.6, 142.4) kg, and had a BMI of 42.8 (39.5, 46.4) kg/m2. Acylated ghrelin, active GLP-1, and total PYY were measured in the fasted state and half-hourly for 2.5h after a standardized meal. We evaluated associations between genotype and appetite hormones in regression analysis controlling for FM and sex. Genotype did not associate with fasting or postprandial (area under curve, AUC) GLP-1 or PYY. Genotype did not associate with fasting acylated ghrelin, but in females with genotype AA, increased FM was associated with higher fasting and postprandial (AUC) acylated ghrelin concentrations relative to genotypes TT (fasting p = 0.025; AUC p = 0.004) and AT (fasting p = 0.002; AUC p < 0.001). This novel finding warrants further investigation.
Collapse
Affiliation(s)
- Ann Kristin Hjelle de Soysa
- Outpatient Obesity Clinic, Clinic of Surgery, St. Olavs hospital–Trondheim University Hospital, Trondheim, Norway
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Mette Langaas
- Department of Mathematical Sciences, Faculty of Information Technology and Electrical Engineering, Norwegian University of Science and Technology, Trondheim, Norway
| | - Valdemar Grill
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
| | - Catia Martins
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Center for Obesity Research and Innovation (ObeCe), Clinic of Surgery, St. Olavs Hospital–Trondheim University Hospital, Trondheim, Norway
| | - Ingrid Løvold Mostad
- Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway
- Department of Nutrition and Speech-Language Therapy, Clinic of Rehabilitation, St. Olavs hospital–Trondheim University Hospital, Trondheim, Norway
| |
Collapse
|
4
|
Masip G, Nielsen DE. Relationships between the Planetary Health Diet Index, its food groups, and polygenic risk of obesity in the CARTaGENE cohort. Nutr Metab (Lond) 2024; 21:116. [PMID: 39741271 DOI: 10.1186/s12986-024-00890-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 12/18/2024] [Indexed: 01/02/2025] Open
Abstract
BACKGROUND The Planetary Health Diet, proposed by the EAT-Lancet Commission, seeks to promote a sustainable and healthy diet for both humans and the environment. However, few studies have investigated relationships between the Planetary Health Diet and the genetic pathway of obesity. The aim of this study was to assess whether adherence to a Planetary Health Diet Index (PHDI) mediated or moderated the genetic susceptibility to obesity. METHODS Participants were 7,037 adults (57% females, aged 55.6 ± 7.7) from the Quebec CARTaGENE Biobank. We constructed a primary polygenic risk score (PRS-Khera) for body mass index (BMI) comprised of ~ 2 million SNPs and utilized a secondary 97 SNPs polygenic risk score (PRS-Locke) for sensitivity analyses. The PHDI was based on 16 food groups. General linear models were conducted to assess main effect associations between the PRSs, the Planetary Health Diet Index (PHDI), and the individual food groups that comprise the PHDI on obesity outcomes. Causal mediation analyses (CMA) were used to evaluate mediation and interaction effects. All models were adjusted for age, sex, genetic ancestry, socio-demographic, and lifestyle variables, including those associated with dietary habits. RESULTS The overall PHDI was inversely associated with BMI (β = - 0.11, 95% confidence interval (CI): - 0.13, - 0.09), waist circumference (WC) (β = - 0.12, 95% CI: - 0.14, - 0.10), and body fat % (β = - 0.10, 95% CI: - 0.12, - 0.08) for all participants, but did not mediate or moderate obesity polygenic risk. Associations between the PRS-Khera and obesity outcomes in all participants were partly mediated by the intake of red meat (mediation effect BMI: 1.72%, p = 0.01; WC: 2.22%, p = 0.01; body fat %: 2.14%, p = 0.01). Moreover, among males, whole grains intake partly mediated the association between the PRS-Khera and outcomes cross-sectionally (BMI: 1.28%, p = 0.03; WC: 1.71%, p = 0.02; body fat %: 2.19%, p = 0.02) and longitudinally (BMI: 3.80%, p = 0.02; WC: 7.38%, p = 0.04), but some observations were attenuated upon correction for multiple comparisons. CONCLUSIONS PHDI adherence was associated with a lower BMI, WC, and body fat % and genetic susceptibility to obesity was partly mediated by the intake of red meat and whole grains. Some components of a plant-based diet could be implicated in mechanisms underlying genetic susceptibility to obesity.
Collapse
Affiliation(s)
- Guiomar Masip
- School of Human Nutrition, McGill University, 21111 Lakeshore Road, Sainte-Anne-de-Bellevue, QC, H9X 3V9, Canada
- GENUD (Growth, Exercise, Nutrition and Development) Research Group, Facultad de Ciencias de la Salud, Universidad de Zaragoza, Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria de Aragón (IISA), Zaragoza, Spain
- Consorcio CIBER, M.P. Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Daiva E Nielsen
- School of Human Nutrition, McGill University, 21111 Lakeshore Road, Sainte-Anne-de-Bellevue, QC, H9X 3V9, Canada.
| |
Collapse
|
5
|
Zhao QG, Song ZT, Ma XL, Xu Q, Bu F, Li K, Zhang L, Pei YF. Human brain proteome-wide association study provides insights into the genetic components of protein abundance in obesity. Int J Obes (Lond) 2024; 48:1603-1612. [PMID: 39025989 DOI: 10.1038/s41366-024-01592-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 07/10/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024]
Abstract
BACKGROUNDS Genome-wide association studies have identified multiple genetic variants associated with obesity. However, most obesity-associated loci were waiting to be translated into new biological insights. Given the critical role of brain in obesity development, we sought to explore whether obesity-associated genetic variants could be mapped to brain protein abundances. METHODS We performed proteome-wide association studies (PWAS) and colocalization analyses to identify genes whose cis-regulated brain protein abundances were associated with obesity-related traits, including body fat percentage, trunk fat percentage, body mass index, visceral adipose tissue, waist circumference, and waist-to-hip ratio. We then assessed the druggability of the identified genes and conducted pathway enrichment analysis to explore their functional relevance. Finally, we evaluated the effects of the significant PWAS genes at the brain transcriptional level. RESULTS By integrating human brain proteomes from discovery (ROSMAP, N = 376) and validation datasets (BANNER, N = 198) with genome-wide summary statistics of obesity-related phenotypes (N ranged from 325,153 to 806,834), we identified 51 genes whose cis-regulated brain protein abundance was associated with obesity. These 51 genes were enriched in 11 metabolic processes, e.g., small molecule metabolic process and metabolic pathways. Fourteen of the 51 genes had high drug repurposing value. Ten of the 51 genes were also associated with obesity at the transcriptome level, suggesting that genetic variants likely confer risk of obesity by regulating mRNA expression and protein abundance of these genes. CONCLUSIONS Our study provides new insights into the genetic component of human brain protein abundance in obesity. The identified proteins represent promising therapeutic targets for future drug development.
Collapse
Affiliation(s)
- Qi-Gang Zhao
- Department of Epidemiology and Biostatistics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, PR China
| | - Zi-Tong Song
- Department of Epidemiology and Biostatistics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, PR China
| | - Xin-Ling Ma
- Department of Epidemiology and Biostatistics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, PR China
| | - Qian Xu
- Department of Epidemiology and Biostatistics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, PR China
| | - Fan Bu
- Department of Epidemiology and Biostatistics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, PR China
| | - Kuan Li
- Department of Epidemiology and Biostatistics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, PR China
| | - Lei Zhang
- Center for Genetic Epidemiology and Genomics, School of Public Health, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, PR China.
| | - Yu-Fang Pei
- Department of Epidemiology and Biostatistics, School of Public Health, Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, MOE Key Laboratory of Geriatric Diseases and Immunology, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, PR China.
| |
Collapse
|
6
|
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
|
7
|
Lahousse L. Overweight and dysanapsis in childhood asthma. Eur Respir J 2024; 64:2401164. [PMID: 39237315 DOI: 10.1183/13993003.01164-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 06/17/2024] [Indexed: 09/07/2024]
Affiliation(s)
- Lies Lahousse
- Department of Bioanalysis, Ghent University, Ghent, Belgium
- Department of Epidemiology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
8
|
Jensen SK, Pedersen CET, Fischer-Rasmussen K, Melgaard ME, Brustad N, Kyvsgaard JN, Vahman N, Schoos AMM, Stokholm J, Chawes B, Eliasen A, Bønnelykke K. Genetic predisposition to high BMI increases risk of early life respiratory infections and episodes of severe wheeze and asthma. Eur Respir J 2024; 64:2400169. [PMID: 38811044 DOI: 10.1183/13993003.00169-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/20/2024] [Indexed: 05/31/2024]
Abstract
BACKGROUND High body mass index (BMI) is an established risk factor for asthma, but the underlying mechanisms remain unclear. OBJECTIVE To increase understanding of the BMI-asthma relationship by studying the association between genetic predisposition to higher BMI and asthma, infections and other asthma traits during childhood. METHODS Data were obtained from the two ongoing Copenhagen Prospective Studies on Asthma in Childhood (COPSAC) mother-child cohorts. Polygenic risk scores for adult BMI were calculated for each child. Replication was done in the large-scale register-based Integrative Psychiatric Research (iPSYCH) cohort using data on hospitalisation for asthma and infections. RESULTS In the COPSAC cohorts (n=974), the adult BMI polygenic risk score was significantly associated with lower respiratory tract infections (incidence rate ratio (IRR) 1.20, 95% CI 1.08-1.33, false discovery rate p-value (pFDR)=0.005) at age 0-3 years and episodes of severe wheeze (IRR 1.30, 95% CI 1.06-1.60, pFDR=0.04) at age 0-6 years. Lower respiratory tract infections partly mediated the association between the adult BMI polygenic risk score and severe wheeze (proportion mediated: 0.59, 95% CI 0.28-2.24, p-value associated with the average causal mediation effect (pACME)=2e-16). In contrast, these associations were not mediated through the child's current BMI and the polygenic risk score was not associated with an asthma diagnosis or reduced lung function up to age 18 years. The associations were replicated in iPSYCH (n=114 283), where the adult BMI polygenic risk score significantly increased the risk of hospitalisations for lower respiratory tract infections and wheeze or asthma throughout childhood to age 18 years. CONCLUSION Children with genetic predisposition to higher BMI had increased risk of lower respiratory tract infections and severe wheeze, independent of the child's current BMI. These results shed further light on the complex relationship between body mass BMI and asthma.
Collapse
Affiliation(s)
- Signe Kjeldgaard Jensen
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Department of Pediatrics, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Casper-Emil Tingskov Pedersen
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Department of Pediatrics, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Kasper Fischer-Rasmussen
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Department of Pediatrics, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Mathias Elsner Melgaard
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Department of Pediatrics, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Nicklas Brustad
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Department of Pediatrics, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Julie Nyholm Kyvsgaard
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Department of Pediatrics, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Pediatrics, Slagelse Hospital, Slagelse, Denmark
| | - Nilo Vahman
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Department of Pediatrics, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Ann-Marie Malby Schoos
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Department of Pediatrics, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Pediatrics, Slagelse Hospital, Slagelse, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jakob Stokholm
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Department of Pediatrics, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Pediatrics, Slagelse Hospital, Slagelse, Denmark
- Section of Microbiology and Fermentation, Department of Food Science, University of Copenhagen, Copenhagen, Denmark
| | - Bo Chawes
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Department of Pediatrics, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anders Eliasen
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Department of Pediatrics, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Pediatrics, Division of Endocrinology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
- Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Shared senior author
| | - Klaus Bønnelykke
- Copenhagen Prospective Studies on Asthma in Childhood (COPSAC), Department of Pediatrics, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Shared senior author
| |
Collapse
|
9
|
Lavoie O, Turmel A, Mattoon P, Desrosiers WJ, Plamondon J, Michael NJ, Caron A. Hypothalamic GABAergic Neurons Expressing Cellular Retinoic Acid Binding Protein 1 (CRABP1) Are Sensitive to Metabolic Status and Liraglutide in Male Mice. Neuroendocrinology 2024; 114:681-697. [PMID: 38631315 PMCID: PMC11232952 DOI: 10.1159/000538716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 03/29/2024] [Indexed: 04/19/2024]
Abstract
INTRODUCTION Owing to their privileged anatomical location, neurons of the arcuate nucleus of the hypothalamus (ARC) play critical roles in sensing and responding to metabolic signals such as leptin and glucagon-like peptide 1 (GLP-1). In addition to the well-known proopiomelanocortin (POMC)- and agouti-related peptide (AgRP)-expressing neurons, subpopulations of GABAergic neurons are emerging as key regulators of energy balance. However, the precise identity of these metabolic neurons is still elusive. Here, we identified and characterized the molecular signature of a novel population of GABAergic neurons of the ARC expressing Cellular retinoic acid binding protein 1 (Crabp1). METHODS Using a combination of immunohistochemistry and in situ hybridization techniques, we investigated the expression of Crabp1 across the mouse brain and characterized the molecular identity of Crabp1ARC neurons. We also determined whether Crabp1ARC neurons are sensitive to fasting, leptin, and GLP1R agonism by assessing cFOS immunoreactivity as a marker of neuronal activity. RESULTS Crabp1ARC neurons represent a novel GABAergic neuronal population robustly enriched in the ARC and are distinct from the prototypical melanocortin neurons. Crabp1ARC neurons overlap with three subpopulations of yet uncharacterized ARC neurons expressing Htr3b, Tbx19, and Tmem215. Notably, Crabp1ARC neurons express receptors for metabolic hormones and their activity is modulated by the nutritional state and GLP1R agonism. CONCLUSION Crabp1ARC neurons represent a novel heterogeneous population of GABAergic neurons sensitive to metabolic status.
Collapse
Affiliation(s)
- Olivier Lavoie
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada
- Quebec Heart and Lung Institute, Quebec City, QC, Canada
| | - Audrey Turmel
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada
- Quebec Heart and Lung Institute, Quebec City, QC, Canada
| | - Paige Mattoon
- Quebec Heart and Lung Institute, Quebec City, QC, Canada
| | | | | | - Natalie Jane Michael
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada
- Quebec Heart and Lung Institute, Quebec City, QC, Canada
| | - Alexandre Caron
- Faculty of Pharmacy, Université Laval, Quebec City, QC, Canada
- Quebec Heart and Lung Institute, Quebec City, QC, Canada
| |
Collapse
|
10
|
Duarte MKRN, Leite-Lais L, Agnez-Lima LF, Maciel BLL, Morais AHDA. Obesity and Nutrigenetics Testing: New Insights. Nutrients 2024; 16:607. [PMID: 38474735 DOI: 10.3390/nu16050607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/09/2024] [Accepted: 02/13/2024] [Indexed: 03/14/2024] Open
Abstract
BACKGROUND Obesity results from interactions between environmental factors, lifestyle, and genetics. In this scenario, nutritional genomics and nutrigenetic tests stand out, with the promise of helping patients avoid or treat obesity. This narrative review investigates whether nutrigenetic tests may help to prevent or treat obesity. Scientific studies in PubMed Science Direct were reviewed, focusing on using nutrigenetic tests in obesity. The work showed that few studies address the use of tools in obesity. However, most of the studies listed reported their beneficial effects in weight loss. Ethical conflicts were also discussed, as in most countries, there are no regulations to standardize these tools, and there needs to be more scientific knowledge for health professionals who interpret them. International Societies, such as the Academy of Nutrition and Dietetics and the Brazilian Association for the Study of Obesity and Metabolic Syndrome, do not recommend nutrigenetic tests to prevent or treat obesity, especially in isolation. Advancing nutrigenetics depends on strengthening three pillars: regulation between countries, scientific evidence with clinical validity, and professional training.
Collapse
Affiliation(s)
| | - Lúcia Leite-Lais
- Department of Cell Biology and Genetics, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil
| | - Lucymara Fassarella Agnez-Lima
- Biochemistry and Molecular Biology Postgraduate Program, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil
- Department of Cell Biology and Genetics, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil
| | - Bruna Leal Lima Maciel
- Department of Nutrition, Health Sciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil
- Postgraduate Program in Nutrition, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil
- Postgraduate Program in Nutrition, Health Sciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil
| | - Ana Heloneida de Araújo Morais
- Biochemistry and Molecular Biology Postgraduate Program, Biosciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil
- Department of Nutrition, Health Sciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil
- Postgraduate Program in Nutrition, Health Sciences Center, Federal University of Rio Grande do Norte, Natal 59078-970, RN, Brazil
| |
Collapse
|
11
|
Ahmetov II, John G, Semenova EA, Hall ECR. Genomic predictors of physical activity and athletic performance. ADVANCES IN GENETICS 2024; 111:311-408. [PMID: 38908902 DOI: 10.1016/bs.adgen.2024.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/24/2024]
Abstract
Physical activity and athletic performance are complex phenotypes influenced by environmental and genetic factors. Recent advances in lifestyle and behavioral genomics led to the discovery of dozens of DNA polymorphisms (variants) associated with physical activity and allowed to use them as genetic instruments in Mendelian randomization studies for identifying the causal links between physical activity and health outcomes. On the other hand, exercise and sports genomics studies are focused on the search for genetic variants associated with athlete status, sports injuries and individual responses to training and supplement use. In this review, the findings of studies investigating genetic markers and their associations with physical activity and athlete status are reported. As of the end of September 2023, a total of 149 variants have been associated with various physical activity traits (of which 42 variants are genome-wide significant) and 253 variants have been linked to athlete status (115 endurance-related, 96 power-related, and 42 strength-related).
Collapse
Affiliation(s)
- Ildus I Ahmetov
- Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom; Sports Genetics Laboratory, St Petersburg Research Institute of Physical Culture, St. Petersburg, Russia; Laboratory of Genetics of Aging and Longevity, Kazan State Medical University, Kazan, Russia; Department of Physical Education, Plekhanov Russian University of Economics, Moscow, Russia.
| | - George John
- Transform Specialist Medical Centre, Dubai, United Arab Emirates
| | - Ekaterina A Semenova
- Department of Molecular Biology and Genetics, Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia; Research Institute of Physical Culture and Sport, Volga Region State University of Physical Culture, Sport and Tourism, Kazan, Russia
| | - Elliott C R Hall
- Faculty of Health Sciences and Sport, University of Stirling, Stirling, United Kingdom
| |
Collapse
|
12
|
Ang MY, Takeuchi F, Kato N. Deciphering the genetic landscape of obesity: a data-driven approach to identifying plausible causal genes and therapeutic targets. J Hum Genet 2023; 68:823-833. [PMID: 37620670 PMCID: PMC10678330 DOI: 10.1038/s10038-023-01189-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 08/08/2023] [Accepted: 08/15/2023] [Indexed: 08/26/2023]
Abstract
OBJECTIVES Genome-wide association studies (GWAS) have successfully revealed numerous susceptibility loci for obesity. However, identifying the causal genes, pathways, and tissues/cell types responsible for these associations remains a challenge, and standardized analysis workflows are lacking. Additionally, due to limited treatment options for obesity, there is a need for the development of new pharmacological therapies. This study aimed to address these issues by performing step-wise utilization of knowledgebase for gene prioritization and assessing the potential relevance of key obesity genes as therapeutic targets. METHODS AND RESULTS First, we generated a list of 28,787 obesity-associated SNPs from the publicly available GWAS dataset (approximately 800,000 individuals in the GIANT meta-analysis). Then, we prioritized 1372 genes with significant in silico evidence against genomic and transcriptomic data, including transcriptionally regulated genes in the brain from transcriptome-wide association studies. In further narrowing down the gene list, we selected key genes, which we found to be useful for the discovery of potential drug seeds as demonstrated in lipid GWAS separately. We thus identified 74 key genes for obesity, which are highly interconnected and enriched in several biological processes that contribute to obesity, including energy expenditure and homeostasis. Of 74 key genes, 37 had not been reported for the pathophysiology of obesity. Finally, by drug-gene interaction analysis, we detected 23 (of 74) key genes that are potential targets for 78 approved and marketed drugs. CONCLUSIONS Our results provide valuable insights into new treatment options for obesity through a data-driven approach that integrates multiple up-to-date knowledgebases.
Collapse
Affiliation(s)
- Mia Yang Ang
- Department of Clinical Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
- Department of Gene Diagnostics and Therapeutics, Medical Genomics Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan.
| | - Fumihiko Takeuchi
- Department of Gene Diagnostics and Therapeutics, Medical Genomics Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Norihiro Kato
- Department of Clinical Genome Informatics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Gene Diagnostics and Therapeutics, Medical Genomics Center, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
13
|
Abstract
Obesity is a common complex trait that elevates the risk for various diseases, including type 2 diabetes and cardiovascular disease. A combination of environmental and genetic factors influences the pathogenesis of obesity. Advances in genomic technologies have driven the identification of multiple genetic loci associated with this disease, ranging from studying severe onset cases to investigating common multifactorial polygenic forms. Additionally, findings from epigenetic analyses of modifications to the genome that do not involve changes to the underlying DNA sequence have emerged as key signatures in the development of obesity. Such modifications can mediate the effects of environmental factors, including diet and lifestyle, on gene expression and clinical presentation. This review outlines what is known about the genetic and epigenetic contributors to obesity susceptibility, along with the albeit limited therapeutic options currently available. Furthermore, we delineate the potential mechanisms of actions through which epigenetic changes can mediate environmental influences and the related opportunities they present for future interventions in the management of obesity.
Collapse
Affiliation(s)
- Khanh Trang
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
| | - Struan F.A. Grant
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
- Division of Diabetes and Endocrinology, Children’s Hospital of Philadelphia, Philadelphia, PA 19104 USA
- Department of Pediatrics, The University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104 USA
- Department of Genetics, University of Pennsylvania, Philadelphia, PA 19104 USA
| |
Collapse
|
14
|
Passeri A, Municchi D, Cavalieri G, Babicola L, Ventura R, Di Segni M. Linking drug and food addiction: an overview of the shared neural circuits and behavioral phenotype. Front Behav Neurosci 2023; 17:1240748. [PMID: 37767338 PMCID: PMC10520727 DOI: 10.3389/fnbeh.2023.1240748] [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: 06/15/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
Despite a lack of agreement on its definition and inclusion as a specific diagnosable disturbance, the food addiction construct is supported by several neurobiological and behavioral clinical and preclinical findings. Recognizing food addiction is critical to understanding how and why it manifests. In this overview, we focused on those as follows: 1. the hyperpalatable food effects in food addiction development; 2. specific brain regions involved in both food and drug addiction; and 3. animal models highlighting commonalities between substance use disorders and food addiction. Although results collected through animal studies emerged from protocols differing in several ways, they clearly highlight commonalities in behavioral manifestations and neurobiological alterations between substance use disorders and food addiction characteristics. To develop improved food addiction models, this heterogeneity should be acknowledged and embraced so that research can systematically investigate the role of specific variables in the development of the different behavioral features of addiction-like behavior in preclinical models.
Collapse
Affiliation(s)
- Alice Passeri
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Psychology and Center “Daniel Bovet”, Sapienza University, Rome, Italy
| | - Diana Municchi
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Psychology and Center “Daniel Bovet”, Sapienza University, Rome, Italy
| | - Giulia Cavalieri
- Department of Psychology and Center “Daniel Bovet”, Sapienza University, Rome, Italy
| | | | - Rossella Ventura
- Department of Psychology and Center “Daniel Bovet”, Sapienza University, Rome, Italy
- IRCCS San Raffaele, Rome, Italy
| | - Matteo Di Segni
- IRCCS Fondazione Santa Lucia, Rome, Italy
- Department of Psychology and Center “Daniel Bovet”, Sapienza University, Rome, Italy
| |
Collapse
|
15
|
Richter M, Widera S, Malz F, Goltermann J, Steinmann L, Kraus A, Enneking V, Meinert S, Repple J, Redlich R, Leehr EJ, Grotegerd D, Dohm K, Kugel H, Bauer J, Arolt V, Dannlowski U, Opel N. Higher body weight-dependent neural activation during reward processing. Brain Imaging Behav 2023; 17:414-424. [PMID: 37012575 PMCID: PMC10435630 DOI: 10.1007/s11682-023-00769-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2023] [Indexed: 04/05/2023]
Abstract
Obesity is associated with alterations in brain structure and function, particularly in areas related to reward processing. Although brain structural investigations have demonstrated a continuous association between higher body weight and reduced gray matter in well-powered samples, functional neuroimaging studies have typically only contrasted individuals from the normal weight and obese body mass index (BMI) ranges with modest sample sizes. It remains unclear, whether the commonly found hyperresponsiveness of the reward circuit can (a) be replicated in well-powered studies and (b) be found as a function of higher body weight even below the threshold of clinical obesity. 383 adults across the weight spectrum underwent functional magnetic resonance imaging during a common card-guessing paradigm simulating monetary reward. Multiple regression was used to investigate the association of BMI and neural activation in the reward circuit. In addition, a one-way ANOVA model comparing three weight groups (normal weight, overweight, obese) was calculated. Higher BMI was associated with higher reward response in the bilateral insula. This association could no longer be found when participants with obesity were excluded from the analysis. The ANOVA revealed higher activation in obese vs. lean, but no difference between lean and overweight participants. The overactivation of reward-related brain areas in obesity is a consistent finding that can be replicated in large samples. In contrast to brain structural aberrations associated with higher body weight, the neurofunctional underpinnings of reward processing in the insula appear to be more pronounced in the higher body weight range.
Collapse
Affiliation(s)
- Maike Richter
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Department of Psychiatry, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany
| | - Sophia Widera
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Franziska Malz
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Janik Goltermann
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Lavinia Steinmann
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Anna Kraus
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Verena Enneking
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Susanne Meinert
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Institute for Translational Neuroscience, University of Münster, Münster, Germany
| | - Jonathan Repple
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Department for Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Frankfurt, Goethe University, Frankfurt, Germany
| | - Ronny Redlich
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
- Department of Psychology, Martin-Luther University of Halle, Halle, Germany
- German Center for Mental Health (DZPG), Jena-Magdeburg-Halle, Germany
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
| | - Elisabeth J Leehr
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Dominik Grotegerd
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Katharina Dohm
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Harald Kugel
- University Clinic for Radiology, University of Münster, Münster, Germany
| | - Jochen Bauer
- University Clinic for Radiology, University of Münster, Münster, Germany
| | - Volker Arolt
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Udo Dannlowski
- Institute for Translational Psychiatry, University of Münster, Münster, Germany
| | - Nils Opel
- Institute for Translational Psychiatry, University of Münster, Münster, Germany.
- Department of Psychiatry, Jena University Hospital/Friedrich-Schiller-University Jena, Jena, Germany.
- German Center for Mental Health (DZPG), Jena-Magdeburg-Halle, Germany.
- Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany.
| |
Collapse
|
16
|
Garske KM, Kar A, Comenho C, Balliu B, Pan DZ, Bhagat YV, Rosenberg G, Koka A, Das SS, Miao Z, Sinsheimer JS, Kaprio J, Pietiläinen KH, Pajukanta P. Increased body mass index is linked to systemic inflammation through altered chromatin co-accessibility in human preadipocytes. Nat Commun 2023; 14:4214. [PMID: 37452040 PMCID: PMC10349101 DOI: 10.1038/s41467-023-39919-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 07/04/2023] [Indexed: 07/18/2023] Open
Abstract
Obesity-induced adipose tissue dysfunction can cause low-grade inflammation and downstream obesity comorbidities. Although preadipocytes may contribute to this pro-inflammatory environment, the underlying mechanisms are unclear. We used human primary preadipocytes from body mass index (BMI) -discordant monozygotic (MZ) twin pairs to generate epigenetic (ATAC-sequence) and transcriptomic (RNA-sequence) data for testing whether increased BMI alters the subnuclear compartmentalization of open chromatin in the twins' preadipocytes, causing downstream inflammation. Here we show that the co-accessibility of open chromatin, i.e. compartmentalization of chromatin activity, is altered in the higher vs lower BMI MZ siblings for a large subset ( ~ 88.5 Mb) of the active subnuclear compartments. Using the UK Biobank we show that variants within these regions contribute to systemic inflammation through interactions with BMI on C-reactive protein. In summary, open chromatin co-accessibility in human preadipocytes is disrupted among the higher BMI siblings, suggesting a mechanism how obesity may lead to inflammation via gene-environment interactions.
Collapse
Affiliation(s)
- Kristina M Garske
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Asha Kar
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Caroline Comenho
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Brunilda Balliu
- Department of Computational Medicine, UCLA, Los Angeles, CA, 90095, USA
| | - David Z Pan
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, 90095, USA
| | - Yash V Bhagat
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Gregory Rosenberg
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Amogha Koka
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Sankha Subhra Das
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Zong Miao
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, 90095, USA
| | - Janet S Sinsheimer
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
- Department of Computational Medicine, UCLA, Los Angeles, CA, 90095, USA
- Bioinformatics Interdepartmental Program, UCLA, Los Angeles, CA, 90095, USA
| | - Jaakko Kaprio
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, 00014, Finland
| | - Kirsi H Pietiläinen
- Obesity Research Unit, Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, 00014, Finland
- Obesity Center, Abdominal Center, Helsinki University Hospital and University of Helsinki, Helsinki, 00014, Finland
| | - Päivi Pajukanta
- Department of Human Genetics, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA.
- Department of Computational Medicine, UCLA, Los Angeles, CA, 90095, USA.
- Institute for Precision Heath, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA.
| |
Collapse
|
17
|
Sumithran P. The Physiological Regulation of Body Fat Mass. Gastroenterol Clin North Am 2023; 52:295-310. [PMID: 37197874 DOI: 10.1016/j.gtc.2023.03.003] [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: 05/19/2023]
Abstract
Disturbances inbody weight and adiposity in both humans and animals are met by compensatory adjustments in energy intake and energy expenditure, suggesting that body weight or fat is regulated. From a clinical viewpoint, this is likely to contribute to the difficulty that many people with obesity have in maintaining weight loss. Finding ways to modify these physiologic responses is likely to improve the long-term success of obesity treatments.
Collapse
Affiliation(s)
- Priya Sumithran
- Department of Medicine (St Vincent's), University of Melbourne, St Vincent's Hospital, Clinical Science Building Level 4, 29 Regent Street, Fitzroy, Victoria 3065, Australia; Department of Endocrinology, Austin Health.
| |
Collapse
|
18
|
Chang X, Chua KY, Ng FL, Wang L, Liu J, Yuan JM, Khor CC, Heng CK, Dorajoo R, Koh WP. Increased BMI and late-life mobility dysfunction; overlap of genetic effects in brain regions. Int J Obes (Lond) 2023; 47:358-364. [PMID: 36788305 DOI: 10.1038/s41366-023-01275-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 02/01/2023] [Accepted: 02/02/2023] [Indexed: 02/16/2023]
Abstract
BACKGROUND How obesity earlier in life impacts upon mobility dysfunctions in late life is not well understood. Pernicious effects of excess weight on the musculoskeletal system and mobility dysfunctions are well-recognized. However, increasingly more data support the link of obesity to overall motor defects that are regulated in the brain. OBJECTIVES To assess the causal relationship between body mass index (BMI) at midlife and performance of the Timed Up-and-Go test (TUG) in late life among a population-based longitudinal cohort of Chinese adults living in Singapore. METHODS We evaluated genetic predispositions for BMI in 8342 participants who were followed up from measurement of BMI at average 53 years, to TUG test (as a functional mobility measure) 20 years later. RESULTS A robust 75.83% of genetically determined BMI effects on late-life TUG scores were mediated through midlife BMI (Pindirect-effect = 9.24 × 10-21). Utilizing Mendelian randomization, we demonstrated a causal effect between BMI and functional mobility in late life (βIVW = 0.180, PIVW = 0.001). Secondary gene enrichment evaluations highlighted down-regulation of genes at BMI risk loci that were correlated with poorer functional mobility in the substantia nigra and amygdala regions as compared to all other tissues. These genes also exhibit differential expression patterns during human brain development. CONCLUSIONS We report a causal effect of obesity on mobility dysfunction. Our findings highlight potential neuronal dysfunctions in regulating predispositions on the causal pathway from obesity to mobility dysfunction.
Collapse
Affiliation(s)
- Xuling Chang
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.,Khoo Teck Puat - National University Children's Medical Institute, National University Health System, Singapore, 119074, Singapore.,Department of Infectious Diseases, The University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, 3000, VIC, Australia
| | - Kevin Yiqiang Chua
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore, 119077, Singapore
| | - Fang Lin Ng
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.,Khoo Teck Puat - National University Children's Medical Institute, National University Health System, Singapore, 119074, Singapore
| | - Ling Wang
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, 138672, Singapore
| | - Jianjun Liu
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, 138672, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Jian-Min Yuan
- Division of Cancer Control and Population Sciences, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, 15232, USA.,Department of Epidemiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Chiea-Chuen Khor
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, 138672, Singapore.,Singapore Eye Research Institute, Singapore National Eye Centre, Singapore, 169856, Singapore
| | - Chew-Kiat Heng
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore. .,Khoo Teck Puat - National University Children's Medical Institute, National University Health System, Singapore, 119074, Singapore.
| | - Rajkumar Dorajoo
- Genome Institute of Singapore, Agency for Science, Technology and Research, Singapore, 138672, Singapore. .,Health Systems and Services Research, Duke-NUS Medical School Singapore, Singapore, 169857, Singapore.
| | - Woon-Puay Koh
- Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117545, Singapore.,Singapore Institute for Clinical Sciences, Agency for Science Technology and Research (A*STAR), Singapore, 117609, Singapore
| |
Collapse
|
19
|
Gerovska D, Araúzo-Bravo MJ. Skeletal Muscles of Sedentary and Physically Active Aged People Have Distinctive Genic Extrachromosomal Circular DNA Profiles. Int J Mol Sci 2023; 24:ijms24032736. [PMID: 36769072 PMCID: PMC9917053 DOI: 10.3390/ijms24032736] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/19/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
To bring new extrachromosomal circular DNA (eccDNA) enrichment technologies closer to the clinic, specifically for screening, early diagnosis, and monitoring of diseases or lifestyle conditions, it is paramount to identify the differential pattern of the genic eccDNA signal between two states. Current studies using short-read sequenced purified eccDNA data are based on absolute numbers of unique eccDNAs per sample or per gene, length distributions, or standard methods for RNA-seq differential analysis. Previous analyses of RNA-seq data found significant transcriptomics difference between sedentary and active life style skeletal muscle (SkM) in young people but very few in old. The first attempt using circulomics data from SkM and blood of aged lifelong sedentary and physically active males found no difference at eccDNA level. To improve the capability of finding differences between circulomics data groups, we designed a computational method to identify Differentially Produced per Gene Circles (DPpGCs) from short-read sequenced purified eccDNA data based on the circular junction, split-read signal, of the eccDNA, and implemented it into a software tool DifCir in Matlab. We employed DifCir to find to the distinctive features of the influence of the physical activity or inactivity in the aged SkM that would have remained undetected by transcriptomics methods. We mapped the data from tissue from SkM and blood from two groups of aged lifelong sedentary and physically active males using Circle_finder and subsequent merging and filtering, to find the number and length distribution of the unique eccDNA. Next, we used DifCir to find up-DPpGCs in the SkM of the sedentary and active groups. We assessed the functional enrichment of the DPpGCs using Disease Gene Network and Gene Set Enrichment Analysis. To find genes that produce eccDNA in a group without comparison with another group, we introduced a method to find Common PpGCs (CPpGCs) and used it to find CPpGCs in the SkM of the sedentary and active group. Finally, we found the eccDNA that carries whole genes. We discovered that the eccDNA in the SkM of the sedentary group is not statistically different from that of physically active aged men in terms of number and length distribution of eccDNA. In contrast, with DifCir we found distinctive gene-associated eccDNA fingerprints. We identified statistically significant up-DPpGCs in the two groups, with the top up-DPpGCs shed by the genes AGBL4, RNF213, DNAH7, MED13, and WWTR1 in the sedentary group, and ZBTB7C, TBCD, ITPR2, and DDX11-AS1 in the active group. The up-DPpGCs in both groups carry mostly gene fragments rather than whole genes. Though the subtle transcriptomics difference, we found RYR1 to be both transcriptionally up-regulated and up-DPpGCs gene in sedentary SkM. DifCir emphasizes the high sensitivity of the circulome compared to the transcriptome to detect the molecular fingerprints of exercise in aged SkM. It allows efficient identification of gene hotspots that excise more eccDNA in a health state or disease compared to a control condition.
Collapse
Affiliation(s)
- Daniela Gerovska
- Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, Calle Doctor Begiristain s/n, 20014 San Sebastian, Spain
- Correspondence: (D.G.); (M.J.A.-B.)
| | - Marcos J. Araúzo-Bravo
- Computational Biology and Systems Biomedicine, Biodonostia Health Research Institute, Calle Doctor Begiristain s/n, 20014 San Sebastian, Spain
- Basque Foundation for Science, IKERBASQUE, Calle María Díaz Harokoa 3, 48013 Bilbao, Spain
- CIBER of Frailty and Healthy Aging (CIBERfes), 28029 Madrid, Spain
- Max Planck Institute for Molecular Biomedicine, Computational Biology and Bioinformatics, Röntgenstr. 20, 48149 Münster, Germany
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of Basque Country (UPV/EHU), 48940 Leioa, Spain
- Correspondence: (D.G.); (M.J.A.-B.)
| |
Collapse
|
20
|
Martingano AJ, Telaak SH, Schopp EM, Fortney C, Dolwick AP, Carnell S, Batheja S, Persky S. Using Educational Videos and Perspective-Taking to Communicate Gene-By-Environment Interaction Concepts about Eating Behavior: Effects on Empathy and Weight Stigma. JOURNAL OF NUTRITION EDUCATION AND BEHAVIOR 2023; 55:55-67. [PMID: 36621267 PMCID: PMC9833839 DOI: 10.1016/j.jneb.2022.09.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 09/13/2022] [Accepted: 09/15/2022] [Indexed: 06/17/2023]
Abstract
OBJECTIVE This study investigated whether education about gene-by-environment interaction (G × E) concepts could improve G × E knowledge and positively affect empathy and weight stigma. DESIGN We conducted a randomized trial using a 2 × 2 between-subjects design. SETTING Online. PARTICIPANTS Five hundred eighty-two American participants from the Prolific platform. INTERVENTION Participants were randomly assigned to watch an educational or a control video. Participants then watched a set of vignette scenarios that depicted what it is like to have a predisposition toward obesogenic eating behaviors from either a first-person or third-person perspective. MAIN OUTCOME MEASURE(S) Participants completed questionnaires measuring G × E knowledge, causal attributions, weight stigma, and empathy postintervention. ANALYSIS Two-by-two between-subjects ANOVAs and exploratory mediation analyses were conducted. RESULTS Participants who watched the educational video demonstrated greater G × E knowledge, reported higher empathy toward the characters in the vignette scenarios and held fewer stigmatizing attitudes (notably blame) toward individuals with higher weight. Exploratory mediation analyses indicated that the educational video led to these positive downstream effects by increasing the extent to which participants attributed genetic causes to eating behaviors. CONCLUSIONS AND IMPLICATIONS Education about G × E causes of eating behaviors can have beneficial downstream effects on attitudes toward people with higher weight.
Collapse
Affiliation(s)
- Alison Jane Martingano
- Social and Behavioral Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Sydney H Telaak
- Social and Behavioral Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Emma M Schopp
- Social and Behavioral Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Christopher Fortney
- Social and Behavioral Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Alexander P Dolwick
- Social and Behavioral Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD
| | - Susan Carnell
- Division of Child and Adolescent Psychiatry, Department of Psychiatry and Behavioral Sciences, Johns Hopkins University, Baltimore, MD
| | - Sapna Batheja
- Department of Food and Nutrition Studies, College of Health and Human Services, George Mason University, Fairfax, VA
| | - Susan Persky
- Social and Behavioral Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
21
|
Next-Generation Sequencing of a Large Gene Panel for Outcome Prediction of Bariatric Surgery in Patients with Severe Obesity. J Clin Med 2022; 11:jcm11247531. [PMID: 36556146 PMCID: PMC9783894 DOI: 10.3390/jcm11247531] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Obesity is a chronic disease in which abnormal deposition of fat threatens health, leading to diabetes, cardiovascular diseases, cancer, and other chronic illnesses. According to the WHO, 19.8% of the adult population in Italy is obese, and the prevalence is higher among men. It is important to know the predisposition of an individual to become obese and to respond to bariatric surgery, the most up-to-date treatment for severe obesity. To this purpose, we developed an NGS gene panel, comprising 72 diagnostic genes and 244 candidate genes, and we sequenced 247 adult obese Italian patients. Eleven deleterious variants in 9 diagnostic genes and 17 deleterious variants in 11 candidate genes were identified. Interestingly, mutations were found in several genes correlated to the Bardet-Biedl syndrome. Then, 25 patients were clinically followed to evaluate their response to bariatric surgery. After a 12-month follow-up, the patients that carried deleterious variants in diagnostic or candidate genes had a reduced weight loss, as compared to the other patients. The NGS-based panel, including diagnostic and candidate genes used in this study, could play a role in evaluating, diagnosing, and managing obese individuals, and may help in predicting the outcome of bariatric surgery.
Collapse
|
22
|
Hwang IS, Hong SB. Association between body mass index and subcortical volume in pre-adolescent children with autism spectrum disorder: An exploratory study. Autism Res 2022; 15:2238-2249. [PMID: 36256577 DOI: 10.1002/aur.2834] [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: 06/11/2022] [Accepted: 10/03/2022] [Indexed: 12/15/2022]
Abstract
Conflicting associations exist between autism spectrum disorder (ASD) and subcortical brain volumes. This study assessed whether obesity might have a confounding influence on associations between ASD and brain subcortical volumes. A comprehensive investigation evaluating the relationship between ASD, obesity, and subcortical structure volumes was conducted. Data obtained included body mass index (BMI) and T1-weighted structural magnetic resonance images for children with and without ASD diagnoses from the Autism Brain Imaging Data Exchange database. Brain subcortical volumes were calculated using vol2Brain software. Hierarchical linear regression analyses were performed to explore the subcortical volumes similarly or differentially associated with BMI in children with or without ASD and examine association and interaction effects regarding ASD and subcortical volume impact on the Social Responsiveness Scale and Vineland Adaptive Behavior Scale (VABS) scores. Bilateral caudate nuclei were smaller in children with ASD than in control participants. Significant interactions were observed between ASD diagnosis and BMI regarding the left caudate, right and left putamen, and right and left ventral diencephalon (DC) volumes (β = -0.384, p = 0.010; β = -0.336, p = 0.030; β = -0.317, p = 0.040; β = 0.322, p = 0.010; β = 0.295, p = 0.021, respectively) and between ASD diagnosis and right and left ventral DC volumes regarding the VABS scores (β = 0.434, p = 0.014; β = 0.495, p = 0.007, respectively). However, each subcortical structure volume included in the ventral DC area could not be measured separately. The results identified subcortical volumes differentially associated with obesity in children with ASD compared with typically developing peers. BMI may need to be considered an important confounder in future research examining brain subcortical volumes within ASD.
Collapse
Affiliation(s)
- In-Seong Hwang
- Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Soon-Beom Hong
- Seoul National University College of Medicine, Seoul, Republic of Korea.,Institute of Human Behavioral Medicine, Seoul National University Medical Research Center, Seoul, Republic of Korea
| |
Collapse
|
23
|
Rakha A, Mehak F, Shabbir MA, Arslan M, Ranjha MMAN, Ahmed W, Socol CT, Rusu AV, Hassoun A, Aadil RM. Insights into the constellating drivers of satiety impacting dietary patterns and lifestyle. Front Nutr 2022; 9:1002619. [PMID: 36225863 PMCID: PMC9549911 DOI: 10.3389/fnut.2022.1002619] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/30/2022] [Indexed: 11/13/2022] Open
Abstract
Food intake and body weight regulation are of special interest for meeting today's lifestyle essential requirements. Since balanced energy intake and expenditure are crucial for healthy living, high levels of energy intake are associated with obesity. Hence, regulation of energy intake occurs through short- and long-term signals as complex central and peripheral physiological signals control food intake. This work aims to explore and compile the main factors influencing satiating efficiency of foods by updating recent knowledge to point out new perspectives on the potential drivers of satiety interfering with food intake regulation. Human internal factors such as genetics, gender, age, nutritional status, gastrointestinal satiety signals, gut enzymes, gastric emptying rate, gut microbiota, individual behavioral response to foods, sleep and circadian rhythms are likely to be important in determining satiety. Besides, the external factors (environmental and behavioral) impacting satiety efficiency are highlighted. Based on mechanisms related to food consumption and dietary patterns several physical, physiological, and psychological factors affect satiety or satiation. A complex network of endocrine and neuroendocrine mechanisms controls the satiety pathways. In response to food intake and other behavioral cues, gut signals enable endocrine systems to target the brain. Intestinal and gastric signals interact with neural pathways in the central nervous system to halt eating or induce satiety. Moreover, complex food composition and structures result in considerable variation in satiety responses for different food groups. A better understanding of foods and factors impacting the efficiency of satiety could be helpful in making smart food choices and dietary recommendations for a healthy lifestyle based on updated scientific evidence.
Collapse
Affiliation(s)
- Allah Rakha
- National Institute of Food Science and Technology, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Fakiha Mehak
- National Institute of Food Science and Technology, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | - Muhammad Asim Shabbir
- National Institute of Food Science and Technology, University of Agriculture Faisalabad, Faisalabad, Pakistan
- *Correspondence: Muhammad Asim Shabbir
| | - Muhammad Arslan
- School of Food and Biological Engineering, Jiangsu University, Zhenjiang, China
| | | | - Waqar Ahmed
- National Institute of Food Science and Technology, University of Agriculture Faisalabad, Faisalabad, Pakistan
| | | | - Alexandru Vasile Rusu
- Life Science Institute, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Cluj-Napoca, Romania
- Faculty of Animal Science and Biotechnology, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, Cluj-Napoca, Romania
- Alexandru Vasile Rusu
| | - Abdo Hassoun
- Univ. Littoral Côte d'Opale, UMRt 1158 BioEcoAgro, USC ANSES, INRAe, Univ. Artois, Univ. Lille, Univ. Picardie Jules Verne, Univ. Liège, Junia, F-62200, Boulogne-sur-Mer, France
- Sustainable AgriFoodtech Innovation & Research (SAFIR), Arras, France
| | - Rana Muhammad Aadil
- National Institute of Food Science and Technology, University of Agriculture Faisalabad, Faisalabad, Pakistan
- Rana Muhammad Aadil
| |
Collapse
|
24
|
Thompson WD, Beaumont RN, Kuang A, Warrington NM, Ji Y, Tyrrell J, Wood AR, Scholtens DM, Knight BA, Evans DM, Lowe Jr WL, Santorelli G, Azad R, Mason D, Hattersley AT, Frayling TM, Yaghootkar H, Borges MC, Lawlor DA, Freathy RM. Fetal alleles predisposing to metabolically favorable adiposity are associated with higher birth weight. Hum Mol Genet 2022; 31:1762-1775. [PMID: 34897462 PMCID: PMC9169452 DOI: 10.1093/hmg/ddab356] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 12/03/2021] [Accepted: 12/07/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Higher birthweight is associated with higher adult body mass index (BMI). Alleles that predispose to greater adult adiposity might act in fetal life to increase fetal growth and birthweight. Whether there are fetal effects of recently identified adult metabolically favorable adiposity alleles on birthweight is unknown. AIM We aimed to test the effect on birthweight of fetal genetic predisposition to higher metabolically favorable adult adiposity and compare that with the effect of fetal genetic predisposition to higher adult BMI. METHODS We used published genome wide association study data (n = upto 406 063) to estimate fetal effects on birthweight (adjusting for maternal genotype) of alleles known to raise metabolically favorable adult adiposity or BMI. We combined summary data across single nucleotide polymorphisms (SNPs) with random effects meta-analyses. We performed weighted linear regression of SNP-birthweight effects against SNP-adult adiposity effects to test for a dose-dependent association. RESULTS Fetal genetic predisposition to higher metabolically favorable adult adiposity and higher adult BMI were both associated with higher birthweight (3 g per effect allele (95% CI: 1-5) averaged over 14 SNPs; P = 0.002; 0.5 g per effect allele (95% CI: 0-1) averaged over 76 SNPs; P = 0.042, respectively). SNPs with greater effects on metabolically favorable adiposity tended to have greater effects on birthweight (R2 = 0.2912, P = 0.027). There was no dose-dependent association for BMI (R2 = -0.0019, P = 0.602). CONCLUSIONS Fetal genetic predisposition to both higher adult metabolically favorable adiposity and BMI is associated with birthweight. Fetal effects of metabolically favorable adiposity-raising alleles on birthweight are modestly proportional to their effects on future adiposity, but those of BMI-raising alleles are not.
Collapse
Affiliation(s)
- William D Thompson
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
| | - Robin N Beaumont
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
| | - Alan Kuang
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Nicole M Warrington
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 2BN, UK
- University of Queensland Diamantina Institute, University of Queensland, Brisbane QLD 4102, Australia
- Department of Public Health and Nursing, NTNU, K.G. Jebsen Center for Genetic Epidemiology, Norwegian University of Science and Technology, Postboks 8905, N-7491, Norway
| | - Yingjie Ji
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
| | - Jessica Tyrrell
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
| | - Andrew R Wood
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
| | - Denise M Scholtens
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Bridget A Knight
- NIHR Exeter Clinical Research Facility, Royal Devon and Exeter NHS Foundation Trust, Exeter EX2 5DW, UK
| | - David M Evans
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 2BN, UK
- University of Queensland Diamantina Institute, University of Queensland, Brisbane QLD 4102, Australia
| | - William L Lowe Jr
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Gillian Santorelli
- Bradford Institute for Health Research, Bradford Royal Infirmary, Duckworth Lane, Bradford BD9 6RJ, UK
| | - Raq Azad
- Department of Biochemistry, Bradford Royal Infirmary, Bradford BD9 6DA, UK
| | - Dan Mason
- Bradford Institute for Health Research, Bradford Royal Infirmary, Duckworth Lane, Bradford BD9 6RJ, UK
| | - Andrew T Hattersley
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
| | - Timothy M Frayling
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
| | - Hanieh Yaghootkar
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
| | - Maria Carolina Borges
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 2BN, UK
- Population Health, Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK
| | - Deborah A Lawlor
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 2BN, UK
- Population Health, Bristol Medical School, University of Bristol, Bristol BS8 2BN, UK
- Bristol NIHR Biomedical Research Centre, Bristol BS8 2BN, UK
| | - Rachel M Freathy
- Institute of Biomedical and Clinical Science, University of Exeter Medical School, University of Exeter, Exeter EX2 5DW, UK
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol BS8 2BN, UK
| |
Collapse
|
25
|
Emerging Roles of FTO in Neuropsychiatric Disorders. BIOMED RESEARCH INTERNATIONAL 2022; 2022:2677312. [PMID: 35528183 PMCID: PMC9071897 DOI: 10.1155/2022/2677312] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 04/11/2022] [Indexed: 12/11/2022]
Abstract
FTO (fat mass and obesity associated) is a recently discovered gene related to obesity and expressed in various tissues of the human body, especially with high expression in the brain. Earlier studies have found that FTO is involved in several biological processes, including brain development and function. In particular, recent studies have found that FTO is a demethylase of N6-methyladenosine (m6A) and it can affect neurological function through the m6A modification of mRNA. At present, a number of studies have shown that FTO is associated with many neuropsychiatric disorders. This paper reviews the discovery, structure, function, and tissue expression of FTO followed by discussing the relationship between FTO and neuropsychiatric diseases. In addition, the potential roles of FTO gene in drug addiction, major depression (MDD), and schizophrenia (SCZ) through regulating m6A modification of dopamine related genes were also highlighted.
Collapse
|
26
|
Szalanczy AM, Key CCC, Woods LCS. Genetic variation in satiety signaling and hypothalamic inflammation: merging fields for the study of obesity. J Nutr Biochem 2022; 101:108928. [PMID: 34936921 PMCID: PMC8959400 DOI: 10.1016/j.jnutbio.2021.108928] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 11/08/2021] [Accepted: 12/01/2021] [Indexed: 02/07/2023]
Abstract
Although obesity has been a longstanding health crisis, the genetic architecture of the disease remains poorly understood. Genome-wide association studies have identified many genomic loci associated with obesity, with genes being enriched in the brain, particularly in the hypothalamus. This points to the role of the central nervous system (CNS) in predisposition to obesity, and we emphasize here several key genes along the satiety signaling pathway involved in genetic susceptibility. Interest has also risen regarding the chronic, low-grade obesity-associated inflammation, with a growing concern toward inflammation in the hypothalamus as a precursor to obesity. Recent studies have found that genetic variation in inflammatory genes play a role in obesity susceptibility, and we highlight here several key genes. Despite the interest in the genetic variants of these pathways individually, there is a lack of research that investigates the relationship between the two. Understanding the interplay between genetic variation in obesity genes enriched in the CNS and inflammation genes will advance our understanding of obesity etiology and heterogeneity, improve genetic risk prediction analyses, and highlight new drug targets for the treatment of obesity. Additionally, this increased knowledge will assist in physician's ability to develop personalized nutrition and medication strategies for combating the obesity epidemic. Though it often seems to present universally, obesity is a highly individual disease, and there remains a need in the field to develop methods to treat at the individual level.
Collapse
|
27
|
Abstract
The prevalence of obesity has tripled over the past four decades, imposing an enormous burden on people's health. Polygenic (or common) obesity and rare, severe, early-onset monogenic obesity are often polarized as distinct diseases. However, gene discovery studies for both forms of obesity show that they have shared genetic and biological underpinnings, pointing to a key role for the brain in the control of body weight. Genome-wide association studies (GWAS) with increasing sample sizes and advances in sequencing technology are the main drivers behind a recent flurry of new discoveries. However, it is the post-GWAS, cross-disciplinary collaborations, which combine new omics technologies and analytical approaches, that have started to facilitate translation of genetic loci into meaningful biology and new avenues for treatment.
Collapse
Affiliation(s)
- Ruth J. F. Loos
- grid.5254.60000 0001 0674 042XNovo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark ,grid.59734.3c0000 0001 0670 2351Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, NY USA ,grid.59734.3c0000 0001 0670 2351Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY USA
| | - Giles S. H. Yeo
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, Wellcome-MRC Institute of Metabolic Science, Addenbrooke’s Hospital, Cambridge, UK
| |
Collapse
|
28
|
Claussnitzer M, Susztak K. Gaining insight into metabolic diseases from human genetic discoveries. Trends Genet 2021; 37:1081-1094. [PMID: 34315631 PMCID: PMC8578350 DOI: 10.1016/j.tig.2021.07.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 06/29/2021] [Accepted: 07/05/2021] [Indexed: 12/30/2022]
Abstract
Human large-scale genetic association studies have identified sequence variations at thousands of genetic risk loci that are more common in patients with diverse metabolic disease compared with healthy controls. While these genetic associations have been replicated in multiple large cohorts and sometimes can explain up to 50% of heritability, the molecular and cellular mechanisms affected by common genetic variation associated with metabolic disease remains mostly unknown. A variety of new genome-wide data types, in conjunction with novel biostatistical and computational analytical methodologies and foundational experimental technologies, are paving the way for a principled approach to systematic variant-to-function (V2F) studies for metabolic diseases, turning associated regions into causal variants, cell types and states of action, effector genes, and cellular and physiological mechanisms. Identification of new target genes and cellular programs for metabolic risk loci will improve mechanistic understanding of disease biology and identification of novel therapeutic strategies.
Collapse
Affiliation(s)
- Melina Claussnitzer
- Beth Israel Deaconess Medical Center, The Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| | - Katalin Susztak
- Department of Medicine and Genetics, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
29
|
Masip G, Foraita R, Silventoinen K, Adan RAH, Ahrens W, De Henauw S, Hebestreit A, Keski-Rahkonen A, Lissner L, Mehlig K, Molnar D, Moreno LA, Pigeot I, Russo P, Veidebaum T, Bogl LH, Kaprio J. The temporal relationship between parental concern of overeating and childhood obesity considering genetic susceptibility: longitudinal results from the IDEFICS/I.Family study. Int J Behav Nutr Phys Act 2021; 18:139. [PMID: 34732214 PMCID: PMC8567680 DOI: 10.1186/s12966-021-01205-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 09/28/2021] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Many genes and molecular pathways are associated with obesity, but the mechanisms from genes to obesity are less well known. Eating behaviors represent a plausible pathway, but because the relationships of eating behaviors and obesity may be bi-directional, it remains challenging to resolve the underlying pathways. A longitudinal approach is needed to assess the contribution of genetic risk during the development of obesity in childhood. In this study we aim to examine the relationships between the polygenic risk score for body mass index (PRS-BMI), parental concern of overeating and obesity indices during childhood. METHODS The IDEFICS/I.Family study is a school-based multicenter pan-European cohort of children observed for 6 years (mean ± SD follow-up 5.8 ± 0.4). Children examined in 2007/2008 (wave 1) (mean ± SD age: 4.4 ± 1.1, range: 2-9 years), in 2009/2010 (wave 2) and in 2013/2014 (wave 3) were included. A total of 5112 children (49% girls) participated at waves 1, 2 and 3. For 2656 children with genome-wide data we constructed a PRS based on 2.1 million single nucleotide polymorphisms. Z-score BMI and z-score waist circumference (WC) were assessed and eating behaviors and relevant confounders were reported by parents via questionnaires. Parental concern of overeating was derived from principal component analyses from an eating behavior questionnaire. RESULTS In cross-lagged models, the prospective associations between z-score obesity indices and parental concern of overeating were bi-directional. In mediation models, the association between the PRS-BMI and parental concern of overeating at wave 3 was mediated by baseline z-BMI (β = 0.16, 95% CI: 0.10, 0.21) and baseline z-WC (β = 0.17, 95% CI: 0.11, 0.23). To a lesser extent, baseline parental concern of overeating also mediated the association between the PRS-BMI and z-BMI at wave 3 (β = 0.10, 95% CI: 0.07, 0.13) and z-WC at wave 3 (β = 0.09, 95% CI: 0.07, 0.12). CONCLUSIONS The findings suggest that the prospective associations between obesity indices and parental concern of overeating are likely bi-directional, but obesity indices have a stronger association with future parental concern of overeating than vice versa. The findings suggest parental concern of overeating as a possible mediator in the genetic susceptibility to obesity and further highlight that other pathways are also involved. A better understanding of the genetic pathways that lead to childhood obesity can help to prevent weight gain. TRIAL REGISTRATION Registry number: ISRCTN62310987 Retrospectively registered 17 September 2018.
Collapse
Affiliation(s)
- Guiomar Masip
- Department of Public Health, University of Helsinki, Helsinki, Finland.
| | - Ronja Foraita
- Leibniz Institute for Prevention Research and Epidemiology - BIPS, Bremen, Germany
| | - Karri Silventoinen
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Population Research Unit, Faculty of Social Sciences, University of Helsinki, Helsinki, Finland
| | - Roger A H Adan
- Department of Translational Neuroscience, UMC Utrecht Brain Center, University Medical Center Utrecht and Utrecht University, Utrecht, the Netherlands
| | - Wolfgang Ahrens
- Leibniz Institute for Prevention Research and Epidemiology - BIPS, Bremen, Germany
- Faculty of Mathematics and Computer Science, University of Bremen, Bremen, Germany
| | - Stefaan De Henauw
- Department of Public Health and Primary Care, Faculty of Medicine and Health Sciences, Ghent University, Ghent, Belgium
| | - Antje Hebestreit
- Leibniz Institute for Prevention Research and Epidemiology - BIPS, Bremen, Germany
| | | | - Lauren Lissner
- School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kirsten Mehlig
- School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Dénés Molnar
- Department of Paediatrics, Medical School, University of Pécs, Pécs, Hungary
| | - Luis A Moreno
- GENUD (Growth, Exercise, Nutrition and Development) Research Group, Faculty of Health Sciences, University of Zaragoza Instituto Agroalimentario de Aragón (IA2), Instituto de Investigación Sanitaria de Aragón, Zaragoza, Spain
- Centro de Investigación Biomédica en Red de Fisiopatología de la Obesidad y Nutrición CIBEROBN, Instituto de Salud Carlos III, Madrid, Spain
| | - Iris Pigeot
- Leibniz Institute for Prevention Research and Epidemiology - BIPS, Bremen, Germany
- Faculty of Mathematics and Computer Science, University of Bremen, Bremen, Germany
| | - Paola Russo
- Institute of Food Sciences, National Research Council, Avellino, Italy
| | - Toomas Veidebaum
- Department of Chronic Diseases, National Institute for Health Development, Tallinn, Estonia
| | - Leonie H Bogl
- Department of Epidemiology, Center for Public Health, Medical University of Vienna, Vienna, Austria
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Jaakko Kaprio
- Department of Public Health, University of Helsinki, Helsinki, Finland
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| |
Collapse
|
30
|
Bouchard C. Genetics of Obesity: What We Have Learned Over Decades of Research. Obesity (Silver Spring) 2021; 29:802-820. [PMID: 33899337 DOI: 10.1002/oby.23116] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 12/14/2022]
Abstract
There is a genetic component to human obesity that accounts for 40% to 50% of the variability in body weight status but that is lower among normal weight individuals (about 30%) and substantially higher in the subpopulation of individuals with obesity and severe obesity (about 60%-80%). The appreciation that heritability varies across classes of BMI represents an important advance. After controlling for BMI, ectopic fat and fat distribution traits are characterized by heritability levels ranging from 30% to 55%. Defects in at least 15 genes are the cause of monogenic obesity cases, resulting mostly from deficiencies in the leptin-melanocortin signaling pathway. Approximately two-thirds of the BMI heritability can be imputed to common DNA variants, whereas low-frequency and rare variants explain the remaining fraction. Diminishing allele effect size is observed as the number of obesity-associated variants expands, with most BMI-increasing or -decreasing alleles contributing only a few grams or less to body weight. Obesity-promoting alleles exert minimal effects in normal weight individuals but have larger effects in individuals with a proneness to obesity, suggesting a higher penetrance; however, it is not known whether these larger effect sizes precede obesity or are caused by an obese state. The obesity genetic risk is conditioned by thousands of DNA variants that make genetically based obesity prevention and treatment a major challenge.
Collapse
Affiliation(s)
- Claude Bouchard
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| |
Collapse
|
31
|
Loos RJF, Burant C, Schur EA. Strategies to Understand the Weight-Reduced State: Genetics and Brain Imaging. Obesity (Silver Spring) 2021; 29 Suppl 1:S39-S50. [PMID: 33759393 PMCID: PMC8500189 DOI: 10.1002/oby.23101] [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: 08/18/2020] [Revised: 12/03/2020] [Accepted: 12/03/2020] [Indexed: 11/09/2022]
Abstract
Most individuals with obesity or overweight have difficulty maintaining weight loss. The weight-reduced state induces changes in many physiological processes that appear to drive weight regain. Here, we review the use of cell biology, genetics, and imaging techniques that are being used to begin understanding why weight regain is the normal response to dieting. As with obesity itself, weight regain has both genetic and environmental drivers. Genetic drivers for "thinness" and "obesity" largely overlap, but there is evidence for specific genetic loci that are different for each of these weight states. There is only limited information regarding the genetics of weight regain. Currently, most genetic loci related to weight point to the central nervous system as the organ responsible for determining the weight set point. Neuroimaging tools have proved useful in studying the contribution of the central nervous system to the weight-reduced state in humans. Neuroimaging technologies fall into three broad categories: functional, connectivity, and structural neuroimaging. Connectivity and structural imaging techniques offer unique opportunities for testing mechanistic hypotheses about changes in brain function or tissue structure in the weight-reduced state.
Collapse
Affiliation(s)
- Ruth J. F. Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Charles Burant
- Department of Internal Medicine, University of Washington, Seattle, Washington, USA
| | - Ellen A. Schur
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
32
|
Wiss DA, Avena N, Gold M. Food Addiction and Psychosocial Adversity: Biological Embedding, Contextual Factors, and Public Health Implications. Nutrients 2020; 12:E3521. [PMID: 33207612 PMCID: PMC7698089 DOI: 10.3390/nu12113521] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 11/12/2020] [Accepted: 11/13/2020] [Indexed: 12/13/2022] Open
Abstract
The role of stress, trauma, and adversity particularly early in life has been identified as a contributing factor in both drug and food addictions. While links between traumatic stress and substance use disorders are well documented, the pathways to food addiction and obesity are less established. This review focuses on psychosocial and neurobiological factors that may increase risk for addiction-like behaviors and ultimately increase BMI over the lifespan. Early childhood and adolescent adversity can induce long-lasting alterations in the glucocorticoid and dopamine systems that lead to increased addiction vulnerability later in life. Allostatic load, the hypothalamic-pituitary-adrenal axis, and emerging data on epigenetics in the context of biological embedding are highlighted. A conceptual model for food addiction is proposed, which integrates data on the biological embedding of adversity as well as upstream psychological, social, and environmental factors. Dietary restraint as a feature of disordered eating is discussed as an important contextual factor related to food addiction. Discussion of various public health and policy considerations are based on the concept that improved knowledge of biopsychosocial mechanisms contributing to food addiction may decrease stigma associated with obesity and disordered eating behavior.
Collapse
Affiliation(s)
- David A. Wiss
- Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA 90095, USA;
| | - Nicole Avena
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA;
- Department of Psychology, Princeton University, Princeton, NJ 08540, USA
| | - Mark Gold
- School of Medicine, Washington University in St. Louis, St. Louis, MO 63130, USA
| |
Collapse
|
33
|
Han HY, Paquet C, Dubé L, Nielsen DE. Diet Quality and Food Prices Modify Associations between Genetic Susceptibility to Obesity and Adiposity Outcomes. Nutrients 2020; 12:E3349. [PMID: 33143186 PMCID: PMC7692602 DOI: 10.3390/nu12113349] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 10/27/2020] [Accepted: 10/28/2020] [Indexed: 11/16/2022] Open
Abstract
The role of the retail food environment in obesity risk is unclear, which may be due in part to the lack of consideration of individual differences in the responsivity to food cues. This cross-sectional investigation geo-temporally linked the CARTaGENE biobank (including genetic, dietary, lifestyle, and anthropometric data) with in-store retail food environment data to examine interactions between a polygenic risk score (PRS) for obesity and (1) diet quality (n = 6807) and (2) in-store retail food measures (n = 3718). The outcomes included adiposity-related measures and diet quality assessed using the 2010 Canadian-adapted Healthy Eating Index. A vegetable:soft drink ratio was constructed for each retail measure to assess the relative healthfulness of exposures. Generalized linear models adjusted for individual and neighborhood socio-demographic factors were used to evaluate main and interactive effects. Diet quality significantly modified the association between polygenic risk of obesity and body mass index, waist circumference, and body fat percent. A significant interaction was also observed between PRS and frequency of price discount of vegetables in relation to soft drinks on waist circumference. These results replicate previous reports of diet moderating polygenic risk of obesity and suggest that prices of low vs. high-energy density foods are an intervention target to address population obesity rates.
Collapse
Affiliation(s)
- Hannah Yang Han
- School of Human Nutrition, McGill University, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada;
| | - Catherine Paquet
- Faculté des Sciences de l’Administration, Université Laval, Québec, QC G1V 0A6, Canada;
- Australian Centre for Precision Health, University of South Australia, Adelaide 5001, Australia
| | - Laurette Dubé
- Desautels Faculty of Management, McGill University, Montreal, QC H3A 1G5, Canada;
| | - Daiva E Nielsen
- School of Human Nutrition, McGill University, Sainte-Anne-de-Bellevue, QC H9X 3V9, Canada;
| |
Collapse
|
34
|
Baron M, Froguel P, Bonnefond A. [Something new in the genetics of monogenic obesity and its insights into pathophysiology]. Med Sci (Paris) 2020; 36:859-865. [PMID: 33026327 DOI: 10.1051/medsci/2020156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Obesity is a complex, multifactorial disorder. About 5% of obese patients actually present with a monogenic form of obesity where only one mutation is sufficient to cause the disease. So far, the genes that have been found to be mutated in these monogenic forms play a key role in the leptin/melanocortin pathway which is mainly active in the hypothalamus and which regulates food intake and energy expenditure. Our laboratory has recently reported a novel monogenic form of obesity due to MRAP2 deficiency where, contrary to previously described monogenic forms of obesity, the carriers presented with hyperglycemia and hypertension in addition to obesity, suggesting that MRAP2 might play a pleiotropic role in metabolic tissues, in addition to its role in brain control of food intake and energy expenditure.
Collapse
Affiliation(s)
- Morgane Baron
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Université de Lille, CHU de Lille, 1 place de Verdun, 59045, France
| | - Philippe Froguel
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Université de Lille, CHU de Lille, 1 place de Verdun, 59045, France - Department of Metabolism, Imperial College London, Londres, W12 0NN, Royaume-Uni
| | - Amélie Bonnefond
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Université de Lille, CHU de Lille, 1 place de Verdun, 59045, France - Department of Metabolism, Imperial College London, Londres, W12 0NN, Royaume-Uni
| |
Collapse
|