1
|
Guasch-Ferré M, Wittenbecher C, Palmnäs M, Ben-Yacov O, Blaak EE, Dahm CC, Fall T, Heitmann BL, Licht TR, Löf M, Loos R, Patel CJ, Quarta C, Redman LM, Segal E, Segata N, Snyder M, Sun Q, Tobias DK, Hu FB, Franks PW, Landberg R, Sargent JL, Merino J. Precision nutrition for cardiometabolic diseases. Nat Med 2025; 31:1444-1453. [PMID: 40307513 DOI: 10.1038/s41591-025-03669-9] [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: 11/15/2024] [Accepted: 03/21/2025] [Indexed: 05/02/2025]
Abstract
Precision nutrition is a vibrant and rapidly evolving field of scientific research and innovation with the potential to deliver health, societal and economic benefits by improving healthcare delivery and policies. Advances in deep phenotyping technologies, digital tools and artificial intelligence have made possible early proof-of-concept research that expands the understanding of within- and between-person variability in responses to diet. These studies illustrate the promise of precision nutrition to complement the traditional 'one size fits all' dietary guidelines, which, while considering broad life-stage and disease-specific nutritional requirements, often lack the granularity to account fully for individual variations in nutritional needs and dietary responses. Despite these developments, however, considerable challenges remain before precision nutrition can be implemented on a broader scale. This Review examines the current state of precision nutrition research, with a focus on its application to reducing the incidence and burden of cardiometabolic diseases. We critically examine the evidence base, explore the potential benefits and discuss the challenges and opportunities ahead.
Collapse
Affiliation(s)
- Marta Guasch-Ferré
- Section of Epidemiology, Department of Public Health, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Clemens Wittenbecher
- Department of Life Sciences, SciLifeLab, Chalmers University of Technology, Gothenburg, Sweden
| | - Marie Palmnäs
- Department of Life Sciences, SciLifeLab, Chalmers University of Technology, Gothenburg, Sweden
| | - Orly Ben-Yacov
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ellen E Blaak
- Department of Human Biology, Maastricht University, Maastricht, the Netherlands
- NUTRIM Institute of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, the Netherlands
| | - Christina C Dahm
- Research Unit for Epidemiology, Department of Public Health, Aarhus University, Aarhus, Denmark
| | - Tove Fall
- Department of Medical Sciences, Molecular Epidemiology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Berit L Heitmann
- Research Unit for Dietary Studies, The Parker Institute, Bispebjerg and Frederiksberg Hospital, The Capital Region, Denmark
- Section for General Medicine, The Department of Public Health, University of Copenhagen, Copenhagen, Denmark
- The Boden Initiative, Charles Perkins Centre, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Tine R Licht
- National Food Institute, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Marie Löf
- Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Ruth Loos
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Chirag J Patel
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Carmelo Quarta
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux, France
| | | | - Eran Segal
- Department of Computer Science and Applied Mathematics, Weizmann Institute of Science, Rehovot, Israel
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy
| | - Michael Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Qi Sun
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Deirdre K Tobias
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Division of Preventive Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Frank B Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Paul W Franks
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Clinical Sciences, Lund University Diabetes Centre, Lund University, Malmö, Sweden
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Oxford, UK
| | - Rikard Landberg
- Department of Life Sciences, SciLifeLab, Chalmers University of Technology, Gothenburg, Sweden
| | - Jennifer L Sargent
- School of Public Health, Imperial College, London, UK
- BabelFisk, Helsingborg, Sweden
| | - Jordi Merino
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
- Diabetes Unit, Endocrine Division, Massachusetts General Hospital, Boston, MA, USA.
- Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
2
|
Wallis NJ, McClellan A, Mörseburg A, Kentistou KA, Jamaluddin A, Dowsett GKC, Schofield E, Morros-Nuevo A, Saeed S, Lam BYH, Sumanasekera NT, Chan J, Kumar SS, Zhang RM, Wainwright JF, Dittmann M, Lakatos G, Rainbow K, Withers D, Bounds R, Ma M, German AJ, Ladlow J, Sargan D, Froguel P, Farooqi IS, Ong KK, Yeo GSH, Tadross JA, Perry JRB, Gorvin CM, Raffan E. Canine genome-wide association study identifies DENND1B as an obesity gene in dogs and humans. Science 2025; 387:eads2145. [PMID: 40048553 DOI: 10.1126/science.ads2145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 01/10/2025] [Indexed: 03/29/2025]
Abstract
Obesity is a heritable disease, but its genetic basis is incompletely understood. Canine population history facilitates trait mapping. We performed a canine genome-wide association study for body condition score-a measure of obesity-in 241 Labrador retrievers. Using a cross-species approach, we showed that canine obesity genes are also associated with rare and common forms of obesity in humans. The lead canine association was within the gene DENN domain containing 1B (DENND1B). Each copy of the alternate allele was associated with ~7.5% greater body fat. We demonstrate a role for this gene in regulating signaling and trafficking of melanocortin 4 receptor, a critical controller of energy homeostasis. Thus, canine genetics identified obesity genes and mechanisms relevant to both dogs and humans.
Collapse
Affiliation(s)
- Natalie J Wallis
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Alyce McClellan
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Alexander Mörseburg
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Katherine A Kentistou
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Aqfan Jamaluddin
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK
| | - Georgina K C Dowsett
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Ellen Schofield
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Anna Morros-Nuevo
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Sadia Saeed
- INSERM UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille, France
- University of Lille, Lille University Hospital, Lille, France
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - Brian Y H Lam
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Natasha T Sumanasekera
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Justine Chan
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Sambhavi S Kumar
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Rey M Zhang
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Jodie F Wainwright
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Marie Dittmann
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Gabriella Lakatos
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Kara Rainbow
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - David Withers
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Rebecca Bounds
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre, Cambridge, UK
| | - Marcella Ma
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Alexander J German
- Institute of Life Course and Medical Sciences and School of Veterinary Science, University of Liverpool, Neston, UK
| | - Jane Ladlow
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - David Sargan
- Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Philippe Froguel
- INSERM UMR 1283, CNRS UMR 8199, European Genomic Institute for Diabetes, Institut Pasteur de Lille, Lille, France
- University of Lille, Lille University Hospital, Lille, France
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
| | - I Sadaf Farooqi
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre, Cambridge, UK
| | - Ken K Ong
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Giles S H Yeo
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- MRC Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - John A Tadross
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- Department of Histopathology and Cambridge Genomics Laboratory, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - John R B Perry
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- MRC Epidemiology Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Caroline M Gorvin
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK
| | - Eleanor Raffan
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
- Metabolic Research Laboratories, Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| |
Collapse
|
3
|
Esbati R, Yazdani O, Simonetti J. Management of Obesity-Related Genetic Disorders. Endocrinol Metab Clin North Am 2025; 54:17-38. [PMID: 39919873 DOI: 10.1016/j.ecl.2024.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2025]
Abstract
Obesity-related genetic disorders are marked by severe, early-onset obesity caused by mutations that disrupt key biological mechanisms regulating hunger, energy balance, and fat storage. These disorders commonly impact systems such as the hypothalamic leptin-melanocortin signaling network, which plays a crucial role in controlling appetite and body weight, mainly through the melanocortin-4 receptor (MC4R) pathway. This review explores current management strategies and emerging therapies for genetic obesity disorders, highlighting the importance of treatment approaches and expanded genetic diagnostics to improve outcomes for affected individuals.
Collapse
Affiliation(s)
- Romina Esbati
- Department of Medicine, Division of Endocrinology, Diabetes and Hypternsion, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Omid Yazdani
- Department of Medicine, Division of Gastroenterology, Hepatology and Endoscopy, Brigham and Women's Hospital, Harvard Medical University, Boston, MA 02115, USA
| | - Juliana Simonetti
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Obesity Medicine Program, University of Utah, Salt Lake City, UT 84108, USA.
| |
Collapse
|
4
|
Yuan Y, Hu X, Guo C, Xu Y, Li S, Wen W, Hu X, Zeng F, Cui W, Chen W, Sun X, Hou N, Wang J, Xiao RP, Zhang X. Reduction of intestinal RIPK1 ameliorates HFD-induced metabolic disorders in female mice. iScience 2025; 28:111906. [PMID: 40028283 PMCID: PMC11869535 DOI: 10.1016/j.isci.2025.111906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 11/27/2024] [Accepted: 01/23/2025] [Indexed: 03/05/2025] Open
Abstract
In modern society, excessive nutrient intake from food is a major factor contributing to the development of a series of metabolic disorders and cardiovascular diseases. Further investigation of the mechanisms underlying nutrient absorption in the intestine will help to better understand and develop preventive or therapeutic strategies. In this study, using receptor-interacting protein kinase 1 (Ripk1) intestine-specific heterozygous knockout mice (Ripk1 IEC+/-) and high-fat diet (HFD)-feeding mouse model, we report that HFD-induced shift in the transcriptional profile of the ileum toward that of the jejunum, characterized by increased expression of jejunal feature genes in the ileum, are attenuated in Ripk1 IEC+/- female mice, but not in males. Accordingly, HFD-induced metabolic disorders, including obesity, impaired glucose tolerance, insulin resistance, and dyslipidemia, are significantly ameliorated in the Ripk1 IEC+/- female mice. These findings demonstrate a new, sex-specific intestinal regulatory mechanism and highlight the critical role of intestinal RIPK1 in regulating HFD-induced metabolic disorders in females.
Collapse
Affiliation(s)
- Ye Yuan
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
- National Biomedical Imaging Center, Peking University, Beijing 100871, China
| | - Xiaomin Hu
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing 100730, China
| | - Chunguang Guo
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Yihua Xu
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Shihan Li
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Wei Wen
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
- PKU-Nanjing Institute of Translational Medicine, Nanjing 211800, China
| | - Xinli Hu
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Fanxin Zeng
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou 635000, China
| | - Weiyi Cui
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Wenli Chen
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Xueting Sun
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Ning Hou
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Jue Wang
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
| | - Rui-Ping Xiao
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
- State Key Laboratory of Biomembrane and Membrane Biotechnology, Peking-Tsinghua Center for Life Sciences, Beijing 100871, China
| | - Xiuqin Zhang
- Institute of Molecular Medicine, College of Future Technology, Peking University, Beijing 100871, China
- Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking University, Beijing 100871, China
- National Biomedical Imaging Center, Peking University, Beijing 100871, China
| |
Collapse
|
5
|
Koskinas KC, Van Craenenbroeck EM, Antoniades C, Blüher M, Gorter TM, Hanssen H, Marx N, McDonagh TA, Mingrone G, Rosengren A, Prescott EB. Obesity and cardiovascular disease: an ESC clinical consensus statement. Eur J Prev Cardiol 2025; 32:184-220. [PMID: 39210708 DOI: 10.1093/eurjpc/zwae279] [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: 05/14/2024] [Revised: 07/08/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Abstract
The global prevalence of obesity has more than doubled over the past four decades, currently affecting more than a billion individuals. Beyond its recognition as a high-risk condition that is causally linked to many chronic illnesses, obesity has been declared a disease per se that results in impaired quality of life and reduced life expectancy. Notably, two-thirds of obesity-related excess mortality is attributable to cardiovascular disease. Despite the increasingly appreciated link between obesity and a broad range of cardiovascular disease manifestations including atherosclerotic disease, heart failure, thromboembolic disease, arrhythmias, and sudden cardiac death, obesity has been underrecognized and sub-optimally addressed compared with other modifiable cardiovascular risk factors. In the view of major repercussions of the obesity epidemic on public health, attention has focused on population-based and personalized approaches to prevent excess weight gain and maintain a healthy body weight from early childhood and throughout adult life, as well as on comprehensive weight loss interventions for persons with established obesity. This clinical consensus statement by the European Society of Cardiology discusses current evidence on the epidemiology and aetiology of obesity; the interplay between obesity, cardiovascular risk factors and cardiac conditions; the clinical management of patients with cardiac disease and obesity; and weight loss strategies including lifestyle changes, interventional procedures, and anti-obesity medications with particular focus on their impact on cardiometabolic risk and cardiac outcomes. The document aims to raise awareness on obesity as a major risk factor and provide guidance for implementing evidence-based practices for its prevention and optimal management within the context of primary and secondary cardiovascular disease prevention.
Collapse
Affiliation(s)
- Konstantinos C Koskinas
- Department of Cardiology, Bern University Hospital-INSELSPITAL, University of Bern, Freiburgstrasse 18, Bern 3010, Switzerland
| | - Emeline M Van Craenenbroeck
- Department of Cardiology, Antwerp University Hospital, Drie Eikenstraat 655, Antwerp 2650, Belgium
- Research group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
| | - Charalambos Antoniades
- Acute Multidisciplinary Imaging and Interventional Centre Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Matthias Blüher
- Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Thomas M Gorter
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Henner Hanssen
- Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Nikolaus Marx
- Department of Internal Medicine I-Cardiology, RWTH Aachen University, Aachen, Germany
| | - Theresa A McDonagh
- Cardiology Department, King's College Hospital, London, UK
- King's College, London, UK
| | - Geltrude Mingrone
- Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, UK
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli & Catholic University, Rome, Italy
| | - Annika Rosengren
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska University Hospital/Ostra, Västra Götaland Region, Gothenburg, Sweden
| | - Eva B Prescott
- Bispebjerg Frederiksberg Hospital, University of Copenhagen, Bispebjerg Bakke 23, Copenhagen 2400, Denmark
| |
Collapse
|
6
|
Dash S. Obesity and Cardiometabolic Disease: Insights From Genetic Studies. Can J Cardiol 2025:S0828-282X(25)00104-7. [PMID: 39920990 DOI: 10.1016/j.cjca.2025.01.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 02/10/2025] Open
Abstract
Obesity is a highly prevalent chronic disease and major driver of both atherosclerotic heart disease and heart failure. Obesity is also a heritable neuronal disease with heritability estimates of up to 70%. In this work I review how common genetic variants, usually with small effect sizes, contribute to the risk for developing obesity and cardiometabolic disease in the majority of people and how this can be further modulated by environmental factors. In some individuals, rare genetic variants with large effect sizes can influence the risk of developing obesity, in some cases in a Mendelian manner. I also address how identification of these rare variants has led to fundamental biologic insights into how satiety and reward are biologic processes, has led to more personalized treatments, and has identified potential novel drug treatments. Biologic insights derived from genetic studies of obesity have also improved our understanding of the causal mediators between obesity and cardiovascular disease. A major limitation of studies to date is that they involved mostly people of European ancestry. Studying more diverse populations will improve our understanding of obesity and cardiometabolic disease. Lessons derived from genetic studies make a compelling case for increasing accessibility to therapies that have sustained efficacy in managing obesity and improving health. This increased knowledge must also inform public health initiatives that will reduce the prevalence of obesity in the coming decades.
Collapse
Affiliation(s)
- Satya Dash
- Department of Medicine, University of Toronto and University Health Network, Toronto, Ontario, Canada.
| |
Collapse
|
7
|
Farooqi S. Understanding the desire for dessert. Science 2025; 387:717-718. [PMID: 39946484 DOI: 10.1126/science.adv4359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Abstract
A neural circuit in mice mediates the preference for high-sugar food after a meal.
Collapse
Affiliation(s)
- Sadaf Farooqi
- Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| |
Collapse
|
8
|
Penninx BW, Lamers F, Jansen R, Berk M, Khandaker GM, De Picker L, Milaneschi Y. Immuno-metabolic depression: from concept to implementation. THE LANCET REGIONAL HEALTH. EUROPE 2025; 48:101166. [PMID: 39801616 PMCID: PMC11721223 DOI: 10.1016/j.lanepe.2024.101166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/21/2024] [Accepted: 11/25/2024] [Indexed: 01/03/2025]
Abstract
Major depressive disorder is a common, disabling mental disorder characterized by extensive etiological and phenotypic heterogeneity. This heterogeneity makes treatment approaches imprecise and often ineffective. Insight into the underlying biological mechanisms underpinning depression and its subtypes may enable more personalized treatments. In this review, we provide an overview of immuno-metabolic depression and illustrate that significant immuno-metabolic dysregulations are present in about 20-30% of people with depression. Such immuno-metabolic depression is characterized by the clustering of 1) atypical, energy-related depressive symptoms such as hypersomnia, fatigue, hyperphagia, and possibly anhedonia, 2) systemic low-grade inflammation with elevated levels of e.g., C-reactive protein, cytokines and glycoprotein acetyls, and 3) metabolic abnormalities involving e.g., obesity, dyslipidaemia, insulin and leptin resistance. Persons with immuno-metabolic depression are at a higher risk for cardiometabolic diseases and seem to respond less well to standard antidepressant treatment. Interventions targeting inflammation, metabolism or lifestyle may be more effective treatment options for individuals with immuno-metabolic depression, in line with principles of precision psychiatry.
Collapse
Affiliation(s)
- Brenda W.J.H. Penninx
- Department of Psychiatry, Amsterdam Public Health and Amsterdam Neuroscience, Amsterdam UMC, Vrije University, Amsterdam, the Netherlands
| | - Femke Lamers
- Department of Psychiatry, Amsterdam Public Health and Amsterdam Neuroscience, Amsterdam UMC, Vrije University, Amsterdam, the Netherlands
| | - Rick Jansen
- Department of Psychiatry, Amsterdam Public Health and Amsterdam Neuroscience, Amsterdam UMC, Vrije University, Amsterdam, the Netherlands
| | - Michael Berk
- Deakin University, IMPACT, The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia
| | - Golam M. Khandaker
- Medical Research Council Integrative Epidemiology Unit, Bristol Medical School, University of Bristol, Bristol, UK
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- National Institute for Health and Care Research Bristol Biomedical Research Centre, United Hospitals Bristol and Weston NHS Foundation Trust, Bristol, UK
- Avon and Wiltshire Mental Health Partnership NHS Trust, Bristol, UK
| | - Livia De Picker
- Collaborative Antwerp Psychiatric Research Institute, Faculty of Health Sciences, University of Antwerp, Antwerp, Belgium
- University Psychiatric Hospital Campus Duffel, Duffel, Belgium
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam Public Health and Amsterdam Neuroscience, Amsterdam UMC, Vrije University, Amsterdam, the Netherlands
| |
Collapse
|
9
|
Argente J, Verge CF, Okorie U, Fennoy I, Kelsey MM, Cokkinias C, Scimia C, Lee HM, Farooqi IS. Setmelanotide in patients aged 2-5 years with rare MC4R pathway-associated obesity (VENTURE): a 1 year, open-label, multicenter, phase 3 trial. Lancet Diabetes Endocrinol 2025; 13:29-37. [PMID: 39549719 DOI: 10.1016/s2213-8587(24)00273-0] [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: 06/25/2024] [Revised: 08/20/2024] [Accepted: 08/20/2024] [Indexed: 11/18/2024]
Abstract
BACKGROUND Setmelanotide, a melanocortin-4 receptor (MC4R) agonist, has been shown to reduce hunger and weight in patients aged 6 years and older with proopiomelanocortin (POMC) deficiency (including biallelic variants in proprotein convertase subtilisin/kexin type 1 [PCSK1]), leptin receptor (LEPR) deficiency, or Bardet-Biedl syndrome (BBS). No approved therapies for patients younger than 6 years old currently exist. The phase 3, open-label VENTURE trial aimed to evaluate the efficacy and safety of setmelanotide in patients aged 2-5 years with POMC or LEPR deficiency or BBS. METHODS This phase 3, open-label, multicentre trial, conducted across six sites in the USA, the UK, Spain, and Australia, enrolled eligible patients aged 2-5 years who had hyperphagia and obesity due to biallelic POMC (including PCSK1) or LEPR variants or genetically confirmed BBS. Open-label subcutaneous setmelanotide was administered once daily for 52 weeks, starting at 0·5 mg with doses increasing every 2 weeks in 0·5 mg increments until reaching the maximum dose based on weight. The co-primary endpoints at week 52 were the percentage of patients reaching a 0·2-point decrease or greater in BMI Z score (a statistical measure used to assess BMI in paediatric patients considering a patient's BMI and comparing it to reference values for the same age and sex) and mean percent change in BMI. Additional endpoints measured safety, hunger, weight-related outcomes, and caregiver burden. The study is registered at ClinicalTrials.gov (NCT04966741) and is complete. FINDINGS Between March 8, 2022, and Sept 18, 2023, 13 patients were screened at the six sites, and 12 patients were enrolled in the study (seven with POMC or LEPR and five with BBS); one patient with BBS was excluded as their BMI was not at the 97th percentile or above. Of the 12 patients enrolled, most were male (seven [58%] vs five [42%] for female) and the mean age was 3·6 years (SD 0·9). 11 patients completed the trial. Ten (83%) of the 12 overall participants reached a 0·2-point reduction or more in BMI Z score per WHO methodology at week 52 (95% CI 58·7-99·8). The mean percent change in BMI from baseline at week 52 was -18% (SD 13) in the overall safety population. Mean percent change in BMI at week 52 was -26% (SD 11) in patients with POMC or LEPR deficiency and -10% (9) in patients with BBS. Mean reductions in secondary endpoints of BMI Z score (3·4 [2·5]) and percent of the BMI 95th percentile (32·5 [22·9]) were seen at Week 52. 91% of caregivers reported that patients were less hungry than at baseline. All adverse events were mild or moderate; skin hyperpigmentation, vomiting, nasopharyngitis, upper respiratory tract infection, and injection site reactions were most common. No serious adverse events or adverse events leading to study discontinuation or death were reported. INTERPRETATION To our knowledge this is the first trial of setmelanotide in patients younger than 6 years old. These results support the benefit of the drug as an early intervention to manage obesity in this population. FUNDING Rhythm Pharmaceuticals.
Collapse
Affiliation(s)
- Jesús Argente
- Department of Paediatrics and Paediatric Endocrinology, University Hospital Niño Jesús, Research Institute La Princesa, Universidad Autónoma de Madrid, Madrid, Spain; CIBER Fisiopatología de la obesidad y nutrición (CIBEROBN), Instituto de Salud Carlos III, Madrid, Spain; IMDEA Food Institute, Madrid, Spain.
| | - Charles F Verge
- Sydney Children's Hospital Randwick and Paediatrics, University of New South Wales, Sydney, NSW, Australia
| | - Uzoma Okorie
- Marshfield Clinic Research Institute, Marshfield, WI, USA
| | - Ilene Fennoy
- Division of Pediatric Endocrinology, Diabetes, and Metabolism, Columbia University Irving Medical Center, New York, NY, USA
| | - Megan M Kelsey
- Department of Pediatrics, University of Colorado School of Medicine and Children's Hospital Colorado, Aurora, CO, USA
| | | | | | | | - I Sadaf Farooqi
- Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|
10
|
Sun Y, Wang W, Li Y, Wang H, Liang L, Wang X, Wang K, Bai W, Luan L, Qin L. Unveiling proteomic targets in the hypothalamus of ovariectomized and estradiol-treated rats: Insights into menopausal syndrome mechanisms. Ann Anat 2025; 257:152341. [PMID: 39326767 DOI: 10.1016/j.aanat.2024.152341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 09/17/2024] [Accepted: 09/20/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Menopausal syndrome profoundly affects the physical and mental health of many women, drawing increasing attention from the medical community. However, its pathogenesis remains unclear. These symptoms are primarily driven by hormonal fluctuation. The hypothalamus, a key regulator of hormonal balance, potentially playing a critical role in the manifestation of menopausal syndrome. METHODS We simulated the low-estrogen menopausal state using ovariectomized rats, confirmed the success of ovariectomy via histological analysis of the uterus and vagina, followed by estrogen treatment. TMT-labeled quantitative proteomics, RTqPCR, targeted proteomics and Western blotting were used to identify differentially expressed proteins and their functions in the hypothalamus under low-estrogen conditions. RESULTS One-way ANOVA (p < 0.05) identified 295 differentially expressed proteins across the sham, ovariectomized and estrogen-treated groups. Post-ovariectomy, 103 differentially expressed proteins were upregulated and 93 were downregulated. Among these, 50 proteins were involved in hormones and neurotransmitters, immunity, metabolism and cardiovascular function. Notably, four proteins-Prkcg, Hsp90ab1, Ywhae, and Gad2-were identified as crucial regulators. CONCLUSIONS This study elucidates the central molecular mechanism of menopausal syndrome through bioinformatics analysis of differentially expressed proteins in the hypothalamus under low-estrogen conditions, providing novel targets for the treatment of related symptoms.
Collapse
Affiliation(s)
- Yanrong Sun
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Wenjuan Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Yao Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266071, China
| | - Hanfei Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Lining Liang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Xiangqiu Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Ke Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Wenpei Bai
- Department of Obstetrics and Gynecology, Beijing Shijitan Hospital Affiliated to Capital Medical University, Beijing 100038, China.
| | - Liju Luan
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.
| | - Lihua Qin
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
11
|
Wang Z, Yang S, Liu L, Mao A, Kan H, Yu F, Ma X, Feng L, Zhou T. The gut microbiota-derived metabolite indole-3-propionic acid enhances leptin sensitivity by targeting STAT3 against diet-induced obesity. Clin Transl Med 2024; 14:e70053. [PMID: 39606796 PMCID: PMC11602751 DOI: 10.1002/ctm2.70053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/21/2024] [Accepted: 09/28/2024] [Indexed: 11/29/2024] Open
Abstract
Obesity is associated with the gut microbiome. Here, we report that gut commensal Clostridia bacteria regulate host energy balance through the tryptophan-derived metabolite indole-3-propionic acid (IPA). IPA acts as an endogenous leptin sensitiser to counteract obesity. Mechanistically, IPA is secreted from the gut into the circulation, and then targets to the STAT3 in the hypothalamic appetite regulation centre, promoting its phosphorylation and nuclear translocation, which enhances the body's response to leptin, and regulates the balance between appetite and energy metabolism. The in vitro pull-down assays involving site-directed mutagenesis demonstrate that Trp623 in the SH2 domain is the key binding site for STAT3-IPA interaction. High-fat diet (HFD), rather than genetic factors, induces excessive secretion of antimicrobial peptides by Paneth cells, inhibiting the growth of Clostridia in the gut and resulting in decreased production of the beneficial metabolite IPA. IPA or Clostridium sporogenes supplement effectively controls weight gain, improves glucose metabolism, and reduces inflammation in DIO mice. IPA fails to achieve such effects in ob/ob mice, while exogenous leptin administration restores the therapeutic effect of IPA. Our study suggests that the IPA-based gut-brain axis regulates host metabolism, and supplementation with microbiome-derived IPA could be a promising intervention strategy for treating obesity.
Collapse
Affiliation(s)
- Zhiwei Wang
- Department of PharmacologyWuxi School of MedicineJiangnan UniversityWuxiChina
| | - Shaying Yang
- Department of PharmacologyWuxi School of MedicineJiangnan UniversityWuxiChina
| | - Liangju Liu
- Department of PharmacologyWuxi School of MedicineJiangnan UniversityWuxiChina
| | - Aiqin Mao
- Department of PharmacologyWuxi School of MedicineJiangnan UniversityWuxiChina
| | - Hao Kan
- Department of PharmacologyWuxi School of MedicineJiangnan UniversityWuxiChina
| | - Fan Yu
- Department of PharmacologyWuxi School of MedicineJiangnan UniversityWuxiChina
| | - Xin Ma
- Department of PharmacologyWuxi School of MedicineJiangnan UniversityWuxiChina
- Medical Basic Research Innovation Center for Gut Microbiota and Chronic DiseasesWuxi School of MedicineJiangnan UniversityWuxiChina
| | - Lei Feng
- Department of PharmacologyWuxi School of MedicineJiangnan UniversityWuxiChina
| | - Tingting Zhou
- Department of PharmacologyWuxi School of MedicineJiangnan UniversityWuxiChina
| |
Collapse
|
12
|
King A, Glaister M, Lawrence K, Nixon J, Pilic L, Mavrommatis Y. A randomised controlled trial to determine the effect of genotype-based personalised diet and physical activity advice for FTO genotype (rs9939609) delivered via email on healthy eating motivation in young adults. NUTR BULL 2024; 49:526-537. [PMID: 39400449 DOI: 10.1111/nbu.12710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 10/15/2024]
Abstract
The prevalence of obesity continues to rise, and public health dietary recommendations are not being adhered to. The transition to higher education is a period of risk for weight gain in young adults and has been demonstrated as a good time to initiate behaviour change. A genotype-based personalised approach to dietary recommendations may motivate young adults to maintain or adopt positive dietary behaviours. The aim of the present study was to determine the efficacy of genotype-based personalised dietary and physical activity advice on healthy eating motivation in young adults. Participants were young adults (n = 153), aged 18-25 years. Baseline measures (participant characteristics, height, weight, body mass index [BMI], body fat percentage [BF%], healthy eating motivation and physical activity) were collected. Participants were genotyped for a SNP in the FTO gene (rs99396090) and randomly allocated (stratified for genotype) to three different groups (1. Genotype-based personalised advice: dietary and physical activity advice based on genotype, BMI and reported physical activity; 2. Non-genotype-based personalised advice: dietary and physical activity advice based on BMI and reported physical activity; 3. Control: no advice). A week after receipt of advice delivered via email, participants completed the healthy eating motivation questionnaire for a second time. Genotype-based personalised dietary advice did not affect healthy eating motivation: when participants were analysed across the whole group (p = 0.417), when analysed according to those informed of a risk or non-risk-associated genotype (p = 0.287), or when analysed according to those with a BMI (>25 kg/m2; p = 0.336) or BF% (male >18%, female >31%; p = 0.387) outside the healthy range. There was also no significant difference in healthy eating motivation at 1-week in the control or non-genotype-based advice groups. Genotype-based personalised advice for the prevention of obesity did not affect healthy eating motivation in this group of healthy, young adults.
Collapse
Affiliation(s)
- Alexandra King
- Faculty of Sport, Technology and Health Sciences, St Mary's University, Twickenham, UK
| | - Mark Glaister
- Faculty of Sport, Technology and Health Sciences, St Mary's University, Twickenham, UK
| | - Kate Lawrence
- Faculty of Sport, Technology and Health Sciences, St Mary's University, Twickenham, UK
| | - Jonathan Nixon
- Faculty of Sport, Technology and Health Sciences, St Mary's University, Twickenham, UK
| | | | | |
Collapse
|
13
|
Winkler TW, Wiegrebe S, Herold JM, Stark KJ, Küchenhoff H, Heid IM. Genetic-by-age interaction analyses on complex traits in UK Biobank and their potential to identify effects on longitudinal trait change. Genome Biol 2024; 25:300. [PMID: 39609904 PMCID: PMC11606088 DOI: 10.1186/s13059-024-03439-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 11/18/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Genome-wide association studies (GWAS) have identified thousands of loci for disease-related human traits in cross-sectional data. However, the impact of age on genetic effects is underacknowledged. Also, identifying genetic effects on longitudinal trait change has been hampered by small sample sizes for longitudinal data. Such effects on deteriorating trait levels over time or disease progression can be clinically relevant. RESULTS Under certain assumptions, we demonstrate analytically that genetic-by-age interaction observed in cross-sectional data can be indicative of genetic association on longitudinal trait change. We propose a 2-stage approach with genome-wide pre-screening for genetic-by-age interaction in cross-sectional data and testing identified variants for longitudinal change in independent longitudinal data. Within UK Biobank cross-sectional data, we analyze 8 complex traits (up to 370,000 individuals). We identify 44 genetic-by-age interactions (7 loci for obesity traits, 26 for pulse pressure, few to none for lipids). Our cross-trait view reveals trait-specificity regarding the proportion of loci with age-modulated effects, which is particularly high for pulse pressure. Testing the 44 variants in longitudinal data (up to 50,000 individuals), we observe significant effects on change for obesity traits (near APOE, TMEM18, TFAP2B) and pulse pressure (near FBN1, IGFBP3; known for implication in arterial stiffness processes). CONCLUSIONS We provide analytical and empirical evidence that cross-sectional genetic-by-age interaction can help pinpoint longitudinal-change effects, when cross-sectional data surpasses longitudinal sample size. Our findings shed light on the distinction between traits that are impacted by age-dependent genetic effects and those that are not.
Collapse
Affiliation(s)
- Thomas W Winkler
- Department of Genetic Epidemiology, University of Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, 93053, Germany.
| | - Simon Wiegrebe
- Department of Genetic Epidemiology, University of Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, 93053, Germany
- Statistical Consulting Unit StaBLab, Department of Statistics, LMU Munich, Geschwister-Scholl-Platz 1, Munich, 80539, Germany
| | - Janina M Herold
- Department of Genetic Epidemiology, University of Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, 93053, Germany
| | - Klaus J Stark
- Department of Genetic Epidemiology, University of Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, 93053, Germany
| | - Helmut Küchenhoff
- Statistical Consulting Unit StaBLab, Department of Statistics, LMU Munich, Geschwister-Scholl-Platz 1, Munich, 80539, Germany
| | - Iris M Heid
- Department of Genetic Epidemiology, University of Regensburg, Franz-Josef-Strauß-Allee 11, Regensburg, 93053, Germany.
| |
Collapse
|
14
|
Bombassaro B, Araujo EP, Velloso LA. The hypothalamus as the central regulator of energy balance and its impact on current and future obesity treatments. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2024; 68:e240082. [PMID: 39876968 PMCID: PMC11771753 DOI: 10.20945/2359-4292-2024-0082] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 06/10/2024] [Indexed: 01/31/2025]
Abstract
The hypothalamus is a master regulator of energy balance in the body. First-order hypothalamic neurons localized in the arcuate nucleus sense systemic signals that indicate the energy stores in the body. Through distinct projections, arcuate nucleus neurons communicate with second-order neurons, which are mostly localized in the paraventricular nucleus and in the lateral hypothalamus. The signals then proceed to third- and fourth-order neurons that activate complex responses aimed at maintaining whole-body energy homeostasis. During the last 30 years, since the identification of leptin in 1994, there has been a great advance in the unveiling of the hypothalamic and extra-hypothalamic neuronal networks that control energy balance. This has contributed to the characterization of the mechanisms by which glucagon-like peptide-1 receptor agonists promote body mass reduction and has opened new windows of opportunity for the development of drugs to treat obesity. This review presents an overview of the mechanisms involved in the hypothalamic regulation of energy balance and discusses how advancements in this field are contributing to the development of new pharmacological strategies to treat obesity.
Collapse
Affiliation(s)
- Bruna Bombassaro
- Universidade de Campinas Centro de Pesquisa em Obesidade e Comorbidades CampinasSP Brasil Centro de Pesquisa em Obesidade e Comorbidades, Universidade de Campinas, Campinas, SP, Brasil
| | - Eliana P Araujo
- Universidade de Campinas Centro de Pesquisa em Obesidade e Comorbidades CampinasSP Brasil Centro de Pesquisa em Obesidade e Comorbidades, Universidade de Campinas, Campinas, SP, Brasil
| | - Licio A Velloso
- Universidade de Campinas Centro de Pesquisa em Obesidade e Comorbidades CampinasSP Brasil Centro de Pesquisa em Obesidade e Comorbidades, Universidade de Campinas, Campinas, SP, Brasil
| |
Collapse
|
15
|
Vourdoumpa A, Paltoglou G, Manou M, Mantzou E, Kassari P, Papadopoulou M, Kolaitis G, Charmandari E. Improvement in Symptoms of Depression and Anxiety and Cardiometabolic Risk Factors in Children and Adolescents with Overweight and Obesity Following the Implementation of a Multidisciplinary Personalized Lifestyle Intervention Program. Nutrients 2024; 16:3710. [PMID: 39519542 PMCID: PMC11547602 DOI: 10.3390/nu16213710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
Background/Objectives: Childhood obesity is one of the most challenging contemporary public health problems. Children and adolescents with obesity experience multiple psychosocial difficulties, such as low self-esteem, depression, anxiety, and behavioral problems, which persist for a long time. The aim of the study was to assess the effect of a multidisciplinary personalized lifestyle intervention for depressive and anxiety symptoms, as evaluated by psychometric questionnaires, and their effect and association with cardiometabolic parameters in children and adolescents with overweight and obesity before and after the intervention. Methods: Six hundred and eleven (n = 611) children and adolescents (mean age ± SE: 10.39 ± 0.10 years; 51.5% females, 46.6% pubertal) were studied prospectively. Subjects were classified as being obese (50.2%), overweight (33.5%), or having a normal BMI (16.2%) according to IOTF criteria. All participants entered a 1-year lifestyle intervention program; laboratory investigations were obtained at the beginning and end of the study and two psychometric questionnaires were completed, the CDI and SCARED, which evaluate symptoms of depression and anxiety, respectively. Results: Following the lifestyle intervention, a significant decrease was noted in anxiety scores in all subjects and in depression scores in youth with obesity, as well as in adolescents with obesity, while females displayed a reduced response to the intervention. Insulin resistance and metabolic syndrome parameters, cortisol, PRL, and LH concentrations were positive predictors for depressive and anxiety symptoms. Conclusions: The implementation of a multidisciplinary personalized lifestyle intervention program in the management of childhood obesity is associated with a significant decrease in cardiometabolic and psychosocial comorbidities in children with and without excess adiposity. The improvement in mental health is likely mediated by an improvement in energy metabolism with subsequent improvement in neuroinflammation owing to lifestyle changes.
Collapse
Affiliation(s)
- Aikaterini Vourdoumpa
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece; (A.V.); (M.M.); (E.M.); (P.K.); (M.P.)
| | - George Paltoglou
- Diabetes and Metabolism Clinic, Second Department of Pediatrics, National and Kapodistrian University of Athens, “P. & A. Kyriakou” Children’s Hospital, 11527 Athens, Greece;
| | - Maria Manou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece; (A.V.); (M.M.); (E.M.); (P.K.); (M.P.)
| | - Emilia Mantzou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece; (A.V.); (M.M.); (E.M.); (P.K.); (M.P.)
| | - Penio Kassari
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece; (A.V.); (M.M.); (E.M.); (P.K.); (M.P.)
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Marina Papadopoulou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece; (A.V.); (M.M.); (E.M.); (P.K.); (M.P.)
| | - Gerasimos Kolaitis
- Department of Child Psychiatry, National and Kapodistrian University of Athens Medical School, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece;
| | - Evangelia Charmandari
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece; (A.V.); (M.M.); (E.M.); (P.K.); (M.P.)
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| |
Collapse
|
16
|
Koskinas KC, Van Craenenbroeck EM, Antoniades C, Blüher M, Gorter TM, Hanssen H, Marx N, McDonagh TA, Mingrone G, Rosengren A, Prescott EB. Obesity and cardiovascular disease: an ESC clinical consensus statement. Eur Heart J 2024; 45:4063-4098. [PMID: 39210706 DOI: 10.1093/eurheartj/ehae508] [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: 05/14/2024] [Revised: 07/08/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
The global prevalence of obesity has more than doubled over the past four decades, currently affecting more than a billion individuals. Beyond its recognition as a high-risk condition that is causally linked to many chronic illnesses, obesity has been declared a disease per se that results in impaired quality of life and reduced life expectancy. Notably, two-thirds of obesity-related excess mortality is attributable to cardiovascular disease. Despite the increasingly appreciated link between obesity and a broad range of cardiovascular disease manifestations including atherosclerotic disease, heart failure, thromboembolic disease, arrhythmias, and sudden cardiac death, obesity has been underrecognized and sub-optimally addressed compared with other modifiable cardiovascular risk factors. In the view of major repercussions of the obesity epidemic on public health, attention has focused on population-based and personalized approaches to prevent excess weight gain and maintain a healthy body weight from early childhood and throughout adult life, as well as on comprehensive weight loss interventions for persons with established obesity. This clinical consensus statement by the European Society of Cardiology discusses current evidence on the epidemiology and aetiology of obesity; the interplay between obesity, cardiovascular risk factors and cardiac conditions; the clinical management of patients with cardiac disease and obesity; and weight loss strategies including lifestyle changes, interventional procedures, and anti-obesity medications with particular focus on their impact on cardiometabolic risk and cardiac outcomes. The document aims to raise awareness on obesity as a major risk factor and provide guidance for implementing evidence-based practices for its prevention and optimal management within the context of primary and secondary cardiovascular disease prevention.
Collapse
Affiliation(s)
- Konstantinos C Koskinas
- Department of Cardiology, Bern University Hospital-INSELSPITAL, University of Bern, Freiburgstrasse 18, Bern 3010, Switzerland
| | - Emeline M Van Craenenbroeck
- Department of Cardiology, Antwerp University Hospital, Drie Eikenstraat 655, Antwerp 2650, Belgium
- Research group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
| | - Charalambos Antoniades
- Acute Multidisciplinary Imaging and Interventional Centre Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Matthias Blüher
- Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Thomas M Gorter
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Henner Hanssen
- Department of Sport, Exercise and Health, University of Basel, Basel, Switzerland
| | - Nikolaus Marx
- Department of Internal Medicine I-Cardiology, RWTH Aachen University, Aachen, Germany
| | - Theresa A McDonagh
- Cardiology Department, King's College Hospital, London, UK
- King's College, London, UK
| | - Geltrude Mingrone
- Cardiovascular and Metabolic Medicine & Sciences, King's College London, London, UK
- Department of Medical and Surgical Sciences, Fondazione Policlinico Universitario A. Gemelli & Catholic University, Rome, Italy
| | - Annika Rosengren
- Department of Molecular and Clinical Medicine, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden
- Sahlgrenska University Hospital/Ostra, Västra Götaland Region, Gothenburg, Sweden
| | - Eva B Prescott
- Bispebjerg Frederiksberg Hospital, University of Copenhagen, Bispebjerg Bakke 23, Copenhagen 2400, Denmark
| |
Collapse
|
17
|
Kar R, Panchali T, Das P, Dutta A, Phoujdar M, Pradhan S. Overview of the therapeutic efficacy of marine fish oil in managing obesity and associated metabolic disorders. Physiol Rep 2024; 12:e70019. [PMID: 39358834 PMCID: PMC11446837 DOI: 10.14814/phy2.70019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/05/2024] [Accepted: 08/14/2024] [Indexed: 10/04/2024] Open
Abstract
In the present scenario, obesity is a challenging health problem and its prevalence along with comorbidities are on the rise around the world. Ingestion of fish becomes trendy in daily meals. Recent research has shown that marine fish oil (FO) (found in tuna, sardines, and mackerel) may offer an alternative method for reducing obesity and problems associated with it. Marine FO rich in long-chain omega-3 polyunsaturated fatty acids (LC n-3 PUFA) and long-chain omega-6 polyunsaturated fatty acids (LC n-6 PUFA) plays an important role in reducing abnormalities associated with the metabolic syndrome and has a variety of disease-fighting properties, including cardioprotective activity, anti-atherosclerotic, anti-obesity, anti-cancer, anti-inflammatory activity. Studies in rodents and humans have indicated that LC n-3 PUFA potentially elicit a number of effects which might be useful for reducing obesity, including suppression of appetite, improvements in circulation, enhanced fat oxidation, energy expenditure, and reduced fat deposition. This review discusses the interplay between inflammation and obesity, and their subsequent regulation via the beneficial role of marine FO, suggesting an alternative dietary strategy to ameliorate obesity and obesity-associated chronic diseases.
Collapse
Affiliation(s)
- Riya Kar
- Biodiversity and Environmental Studies Research CenterMidnapore City College, affiliated to Vidyasagar UniversityMidnaporeWest BengalIndia
- Central Research Laboratory, Department of Paramedical and Allied Health SciencesMidnapore City CollegeMidnaporeWest BengalIndia
| | - Titli Panchali
- Biodiversity and Environmental Studies Research CenterMidnapore City College, affiliated to Vidyasagar UniversityMidnaporeWest BengalIndia
- Central Research Laboratory, Department of Paramedical and Allied Health SciencesMidnapore City CollegeMidnaporeWest BengalIndia
| | - Pipika Das
- Biodiversity and Environmental Studies Research CenterMidnapore City College, affiliated to Vidyasagar UniversityMidnaporeWest BengalIndia
- Central Research Laboratory, Department of Paramedical and Allied Health SciencesMidnapore City CollegeMidnaporeWest BengalIndia
| | - Ananya Dutta
- Biodiversity and Environmental Studies Research CenterMidnapore City College, affiliated to Vidyasagar UniversityMidnaporeWest BengalIndia
- Central Research Laboratory, Department of Paramedical and Allied Health SciencesMidnapore City CollegeMidnaporeWest BengalIndia
| | - Manisha Phoujdar
- Biodiversity and Environmental Studies Research CenterMidnapore City College, affiliated to Vidyasagar UniversityMidnaporeWest BengalIndia
- Central Research Laboratory, Department of Paramedical and Allied Health SciencesMidnapore City CollegeMidnaporeWest BengalIndia
| | - Shrabani Pradhan
- Central Research Laboratory, Department of Paramedical and Allied Health SciencesMidnapore City CollegeMidnaporeWest BengalIndia
| |
Collapse
|
18
|
Qamar S, Mallik R, Makaronidis J. Setmelanotide: A Melanocortin-4 Receptor Agonist for the Treatment of Severe Obesity Due to Hypothalamic Dysfunction. TOUCHREVIEWS IN ENDOCRINOLOGY 2024; 20:62-71. [PMID: 39526054 PMCID: PMC11548362 DOI: 10.17925/ee.2024.20.2.9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/27/2023] [Indexed: 11/16/2024]
Abstract
Obesity is a silent global pandemic. It is a condition associated with multiple risk factors and adverse outcomes that arise from the intertwined relationship between environmental factors and genetics. The genetic factors that cause phenotypic expression are variable. Monogenic obesity is a severe early-onset and rarer form of obesity, which presents with co-morbidities such as abnormal feeding behaviour. Monogenic obesity causes impaired weight regulation in the hypothalamus due to defects in the leptin-melanocortin signalling pathway. The emergence of a new therapeutic treatment, the melanocortin-4 receptor agonist setmelanotide (originally RM-493), has represented a breakthrough in the management of monogenic obesity and has raised hope in managing complex obesity. This review provides an overview of the setmelanotide trials that have taken place, as well as its mechanism of action, side effects and weight loss outcomes that led to its approval in the treatment of pro-opiomelanocortin (POMC) deficiency and proprotein convertase subtilisin/kexin type 1 (PCSK1) deficiency. It also explores setmelanotide's role in other genetic forms of obesity, such as hypothalamic obesity, Prader-Willi syndrome, Alström syndrome and other rare genetic conditions that are being investigated. This review aims to help to understand the pathophysiology of genetic obesity and aid in future treatment options for people with severe, complex genetic obesity.
Collapse
Affiliation(s)
- Sulmaaz Qamar
- Centre for Obesity Research, Rayne Institute, Department of Medicine, University College London, London, UK
- UCLH Bariatric Centre for Weight Management and Metabolic Surgery, University College London Hospital, London, UK
- National Institute of Health Research, UCLH Biomedical Research Centre, London, UK
| | - Ritwika Mallik
- Centre for Obesity Research, Rayne Institute, Department of Medicine, University College London, London, UK
- UCLH Bariatric Centre for Weight Management and Metabolic Surgery, University College London Hospital, London, UK
- National Institute of Health Research, UCLH Biomedical Research Centre, London, UK
| | - Janine Makaronidis
- Centre for Obesity Research, Rayne Institute, Department of Medicine, University College London, London, UK
- UCLH Bariatric Centre for Weight Management and Metabolic Surgery, University College London Hospital, London, UK
- National Institute of Health Research, UCLH Biomedical Research Centre, London, UK
- Department of Diabetes and Metabolism, Barts Health NHS Trust, London, UK
| |
Collapse
|
19
|
Dash S. Opportunities to optimize lifestyle interventions in combination with glucagon-like peptide-1-based therapy. Diabetes Obes Metab 2024; 26 Suppl 4:3-15. [PMID: 39157881 DOI: 10.1111/dom.15829] [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: 04/28/2024] [Revised: 06/28/2024] [Accepted: 07/10/2024] [Indexed: 08/20/2024]
Abstract
Obesity is a chronic multi-system disease and major driver of type 2 diabetes and cardiometabolic disease. Nutritional interventions form the cornerstone of obesity and type 2 diabetes management. Some interventions such as Mediterranean diet can reduce incident cardiovascular disease, probably independently of weight loss. Weight loss of 5% or greater can improve many adiposity-related comorbidities. Although this can be achieved with lifestyle intervention, it is often difficult to sustain in the longer term due to adaptive endocrine changes. In recent years glucagon-like-peptide-1 receptor agonists (GLP-1RAs) have emerged as effective treatments for both type 2 diabetes and obesity. Newer GLP-1RAs can achieve average weight loss of 15% or greater and improve cardiometabolic health. There is heterogeneity in the weight loss response to GLP-1RAs, with a substantial number of patients unable to achieve 5% or greater weight. Weight loss, on average, is lower in older adults, male patients and people with type 2 diabetes. Mechanistic studies are needed to understand the aetiology of this variable response. Gastrointestinal side effects leading to medication discontinuation are a concern with GLP-1RA treatment, based on real-world data. With weight loss of 20% or higher with newer GLP-1RAs, nutritional deficiency and sarcopenia are also potential concerns. Lifestyle interventions that may potentially mitigate the side effects of GLP-1RA treatment and enhance weight loss are discussed here. The efficacy of such interventions awaits confirmation with well-designed randomized controlled trials.
Collapse
Affiliation(s)
- Satya Dash
- Division of Endocrinology, University Health Network & University of Toronto, Toronto General Hospital, Toronto, Ontario, Canada
| |
Collapse
|
20
|
Farooqi IS, Xu Y. Translational potential of mouse models of human metabolic disease. Cell 2024; 187:4129-4143. [PMID: 39067442 DOI: 10.1016/j.cell.2024.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/05/2024] [Accepted: 07/05/2024] [Indexed: 07/30/2024]
Abstract
Obesity causes significant morbidity and mortality globally. Research in the last three decades has delivered a step-change in our understanding of the fundamental mechanisms that regulate energy homeostasis, building on foundational discoveries in mouse models of metabolic disease. However, not all findings made in rodents have translated to humans, hampering drug discovery in this field. Here, we review how studies in mice and humans have informed our current framework for understanding energy homeostasis, discuss their challenges and limitations, and offer a perspective on how human studies may play an increasingly important role in the discovery of disease mechanisms and identification of therapeutic targets in the future.
Collapse
Affiliation(s)
- I Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, Cambridge, UK.
| | - Yong Xu
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Department of Molecular and Cellular Biology and Department of Medicine, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
21
|
Powell DR, Doree DD, Shadoan MK, Platt KA, Brommage R, Vogel P, Revelli JP. Mice Lacking Mrs2 Magnesium Transporter are Hypophagic and Thin When Maintained on a High-Fat Diet. Endocrinology 2024; 165:bqae072. [PMID: 38878275 DOI: 10.1210/endocr/bqae072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Indexed: 07/05/2024]
Abstract
Genes regulating body fat are shared with high fidelity by mice and humans, indicating that mouse knockout (KO) phenotyping might identify valuable antiobesity drug targets. Male Mrs2 magnesium transporter (Mrs2) KO mice were recently reported as thin when fed a high-fat diet (HFD). They also exhibited increased energy expenditure (EE)/body weight and had beiged adipocytes that, along with isolated hepatocytes, demonstrated increased oxygen consumption, suggesting that increased EE drove the thin phenotype. Here we provide our data on these and additional assays in Mrs2 KO mice. We generated Mrs2 KO mice by homologous recombination. HFD-fed male and female Mrs2 KO mice had significantly less body fat, measured by quantitative magnetic resonance, than wild-type (WT) littermates. HFD-fed Mrs2 KO mice did not demonstrate increased EE by indirect calorimetry and could not maintain body temperature at 4 °C, consistent with their decreased brown adipose tissue stores but despite increased beige white adipose tissue. Instead, when provided a choice between HFD and low-fat diet (LFD), Mrs2 KO mice showed a significant 15% decrease in total energy intake resulting from significantly lower HFD intake that offset numerically increased LFD intake. Food restriction studies performed using WT mice suggested that this decrease in energy intake could explain the loss of body fat. Oral glucose tolerance test studies revealed significantly improved insulin sensitivity in Mrs2 KO mice. We conclude that HFD-fed Mrs2 KO mice are thin with improved insulin sensitivity, and that this favorable metabolic phenotype is driven by hypophagia. Further evaluation is warranted to determine the suitability of MRS2 as a drug target for antiobesity therapeutics.
Collapse
Affiliation(s)
| | - Deon D Doree
- Lexicon Pharmaceuticals, The Woodlands, TX 77381, USA
| | | | | | | | - Peter Vogel
- Lexicon Pharmaceuticals, The Woodlands, TX 77381, USA
| | | |
Collapse
|
22
|
Davis MG, Sanders BD. Updates in Medical and Surgical Weight Loss. J Midwifery Womens Health 2024; 69:414-421. [PMID: 38831484 DOI: 10.1111/jmwh.13652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 04/24/2024] [Indexed: 06/05/2024]
Abstract
The number of individuals with obesity is at an all-time high, and the rate of obesity continues to climb each year. Obesity is a chronic disease with widespread effects throughout the body. Midwives and perinatal care providers need an understanding of the etiology, pathophysiology, and interventions for obesity. A review of evidence-based diet and lifestyle modifications, medications, and surgical procedures is presented.
Collapse
Affiliation(s)
- Melissa G Davis
- Vanderbilt University School of Nursing and Vanderbilt University Medical Center, Nashville, Tennessee
| | - Bethany D Sanders
- Vanderbilt University School of Nursing and Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
23
|
Cifuentes L, Ghusn W, Campos A, Bublitz JT, Hurtado MD, Olson J, Acosta A. Leptin-Melanocortin pathway variants and gastric emptying in patients with obesity. Neurogastroenterol Motil 2024; 36:e14764. [PMID: 38361111 PMCID: PMC11042991 DOI: 10.1111/nmo.14764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/16/2024] [Accepted: 02/02/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND Accelerated gastric emptying (GE) is a trait seen in obesity. Mutations in the hypothalamic leptin-melanocortin 4 receptor (Leptin-MC4R) pathway have been associated with obesity. We sought to investigate the association of leptin-MC4R pathway variants and GE in patients with obesity. METHODS This is a cross-sectional study of patients with a history of severe obesity that were genotyped and completed a GE test by scintigraphy. We evaluated the percentage of GE (GE %) at 2 and 4 h between both groups using ANCOVA with weight and sex as covariates. We subdivide patients into carriers based on the location of the identified variants (i.e., upstream or downstream of the Leptin-MC4R pathway) and compared them with noncarriers using ANOVA. Results are presented as mean and standard deviation (± SD). KEY RESULTS A total of 95 patients; nine carriers (67% females; 39.78 ± 12.33 years; BMI: 49.14 ± 12.96 kg/m2) and 86 noncarriers (87% female; 49.98 ± 13.74 years; BMI: 40.75 ± 6.29 kg/m2) were included. At 2 and 4 h, carriers had a delayed GE when compared noncarriers (p = 0.03 and p = 0.005, respectively). In carriers, when compared upstream carriers vs. downstream carriers vs. noncarriers by location there was a significant difference in GE among groups at 2 h and at 4 h (p = 0.02 and p = 0.01, respectively). CONCLUSIONS & INFERENCES Carriers of heterozygous variants in the Leptin-MC4R pathway had a delayed GE compared to noncarriers. These findings point the important relationship between the Leptin-MC4R pathway and gastric motility.
Collapse
Affiliation(s)
- Lizeth Cifuentes
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Wissam Ghusn
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Alejandro Campos
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Joshua T. Bublitz
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Maria Daniela Hurtado
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Janet Olson
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Andres Acosta
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
24
|
Abdisa KB, Szerdahelyi E, Molnár MA, Friedrich L, Lakner Z, Koris A, Toth A, Nath A. Metabolic Syndrome and Biotherapeutic Activity of Dairy (Cow and Buffalo) Milk Proteins and Peptides: Fast Food-Induced Obesity Perspective-A Narrative Review. Biomolecules 2024; 14:478. [PMID: 38672494 PMCID: PMC11048494 DOI: 10.3390/biom14040478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/30/2024] [Accepted: 04/08/2024] [Indexed: 04/28/2024] Open
Abstract
Metabolic syndrome (MS) is defined by the outcome of interconnected metabolic factors that directly increase the prevalence of obesity and other metabolic diseases. Currently, obesity is considered one of the most relevant topics of discussion because an epidemic heave of the incidence of obesity in both developing and underdeveloped countries has been reached. According to the World Obesity Atlas 2023 report, 38% of the world population are presently either obese or overweight. One of the causes of obesity is an imbalance of energy intake and energy expenditure, where nutritional imbalance due to consumption of high-calorie fast foods play a pivotal role. The dynamic interactions among different risk factors of obesity are highly complex; however, the underpinnings of hyperglycemia and dyslipidemia for obesity incidence are recognized. Fast foods, primarily composed of soluble carbohydrates, non-nutritive artificial sweeteners, saturated fats, and complexes of macronutrients (protein-carbohydrate, starch-lipid, starch-lipid-protein) provide high metabolic calories. Several experimental studies have pointed out that dairy proteins and peptides may modulate the activities of risk factors of obesity. To justify the results precisely, peptides from dairy milk proteins were synthesized under in vitro conditions and their contributions to biomarkers of obesity were assessed. Comprehensive information about the impact of proteins and peptides from dairy milks on fast food-induced obesity is presented in this narrative review article.
Collapse
Affiliation(s)
- Kenbon Beyene Abdisa
- Department of Food Process Engineering, Institute of Food Science and Technology, Hungarian University of Agriculture and Life Sciences, Ménesi út 44, HU-1118 Budapest, Hungary; (K.B.A.)
| | - Emőke Szerdahelyi
- Department of Nutrition, Institute of Food Science and Technology, Hungarian University of Agriculture and Life Sciences, Somlói út 14-16, HU-1118 Budapest, Hungary;
| | - Máté András Molnár
- Department of Food Process Engineering, Institute of Food Science and Technology, Hungarian University of Agriculture and Life Sciences, Ménesi út 44, HU-1118 Budapest, Hungary; (K.B.A.)
| | - László Friedrich
- Department of Refrigeration and Livestock Product Technology, Institute of Food Science and Technology, Hungarian University of Agriculture and Life Sciences, Ménesi út 43-45, HU-1118 Budapest, Hungary
| | - Zoltán Lakner
- Department of Agricultural Business and Economics, Institute of Agricultural and Food Economics, Hungarian University of Agriculture and Life Sciences, Villányi út 29-43, HU-1118 Budapest, Hungary
| | - András Koris
- Department of Food Process Engineering, Institute of Food Science and Technology, Hungarian University of Agriculture and Life Sciences, Ménesi út 44, HU-1118 Budapest, Hungary; (K.B.A.)
| | - Attila Toth
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Móricz Zsigmond út 22, HU-4032 Debrecen, Hungary
| | - Arijit Nath
- Department of Food Process Engineering, Institute of Food Science and Technology, Hungarian University of Agriculture and Life Sciences, Ménesi út 44, HU-1118 Budapest, Hungary; (K.B.A.)
| |
Collapse
|
25
|
Wang ZY, Qu YF, Yu TM, Liu ZL, Cheng YG, Zhong MW, Hu SY. Novel subtype of obesity influencing the outcomes of sleeve gastrectomy: Familial aggregation of obesity. World J Gastroenterol 2024; 30:1887-1898. [PMID: 38659480 PMCID: PMC11036498 DOI: 10.3748/wjg.v30.i13.1887] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/07/2024] [Accepted: 03/14/2024] [Indexed: 04/03/2024] Open
Abstract
BACKGROUND Differences in the preoperative characteristics and weight loss outcomes after sleeve gastrectomy (SG) between patients with familial aggregation of obesity (FAO) and patients with sporadic obesity (SO) have not been elucidated. AIM To explore the impact of SG on weight loss and the alleviation of obesity-related comorbidities in individuals with FAO. METHODS A total of 193 patients with obesity who underwent SG were selected. Patients with FAO/SO were matched 1:1 by propensity score matching and were categorized into 4 groups based on the number of first-degree relatives with obesity (1SO vs 1FAO, 2SO vs 2FAO). The baseline characteristics, weight loss outcomes, prevalence of obesity-related comorbidities and incidence of major surgery-related complications were compared between groups. RESULTS We defined FAO as the presence of two or more first-degree relatives with obesity. Patients with FAO did not initially show significant differences in baseline data, short-term postoperative weight loss, or obesity-related comorbidities when compared to patients with SO preoperatively. However, distinctions between the two groups became evident at the two-year mark, with statistically significant differences in both percentage of total weight loss (P = 0.006) and percentage of excess weight loss (P < 0.001). The FAO group exhibited weaker remission of type 2 diabetes mellitus (T2DM) (P = 0.031), hyperlipidemia (P = 0.012), and non-alcoholic fatty liver disease (NAFLD) (P = 0.003) as well as a lower incidence of acid reflux (P = 0.038). CONCLUSION FAO patients is associated with decreased mid-to-long-term weight loss outcomes; the alleviation of T2DM, hyperlipidemia and NAFLD; and decreased incidence of acid reflux postoperatively.
Collapse
Affiliation(s)
- Ze-Yu Wang
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan 250000, Shandong Province, China
- Department of Postgraduate, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250000, Shandong Province, China
| | - Yun-Fei Qu
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan 250000, Shandong Province, China
- Department of Postgraduate, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan 250000, Shandong Province, China
| | - Tian-Ming Yu
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine Shandong University, Jinan 250000, Shandong Province, China
| | - Zeng-Lin Liu
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan 250000, Shandong Province, China
- Department of General Surgery, Shandong Provincial Qianfoshan Hospital, Cheeloo College of Medicine Shandong University, Jinan 250000, Shandong Province, China
| | - Yu-Gang Cheng
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan 250000, Shandong Province, China
| | - Ming-Wei Zhong
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan 250000, Shandong Province, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Lunan Pharmaceutical Group Co. Ltd., Linyi 276005, Shandong Province, China
| | - San-Yuan Hu
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan 250000, Shandong Province, China
| |
Collapse
|
26
|
Supti DA, Akter F, Rahman MI, Munim MA, Tonmoy MIQ, Tarin RJ, Afroz S, Reza HA, Yeasmin R, Alam MR, Hossain MS. Meta-analysis investigating the impact of the LEPR rs1137101 (A>G) polymorphism on obesity risk in Asian and Caucasian ethnicities. Heliyon 2024; 10:e27213. [PMID: 38496879 PMCID: PMC10944198 DOI: 10.1016/j.heliyon.2024.e27213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 12/12/2023] [Accepted: 02/26/2024] [Indexed: 03/19/2024] Open
Abstract
Obesity is a chronic condition which is identified by the buildup of excess body fat caused by a combination of various factors, including genetic predisposition and lifestyle choices. rs1137101 (A > G) polymorphism in the CHR1 domain of LEPR protein linked to different diseases including obesity. Nevertheless, the connection between this polymorphism and the likelihood of developing obesity has not been determined definitively. Therefore, a meta-analysis was conducted to assess the relationship between rs1137101 and the risk of obesity. The meta-analysis included all studies meeting pre-defined criteria, found through searching databases up until February 2023. A combined odds ratio with a 95% confidence interval was estimated as overall and in continent subgroups for homozygous, heterozygous, recessive, dominant and allelic models using the fixed or the random-effects model. The meta-analysis identified 39 eligible studies with cases and controls (6099 cases/6711 controls) in 38 articles under different ethnic backgrounds. The results indicated a significant relationship between rs1137101 and the likelihood of developing obesity in each of the genetic models [the homozygous model (GG vs. AA: 95% Confidence Interval = 1.12-1.73, Odds Ratio = 1.39, P value = 0.003); the heterozygous model (AG vs. AA: 95% Confidence Interval = 1.07-1.42, Odds Ratio = 1.23, P value = 0.005); the dominant model (AG/GG vs AA: 95% Confidence Interval = 1.10-1.49, Odds Ratio = 1.28, P value = 0.001); the recessive model (GG vs AA/AG: 95% Confidence Interval = 1.02-1.45, Odds Ratio = 1.21, P value = 0.03); and the allelic model (G vs A; 95% Confidence Interval = 1.07-1.33, Odds Ratio = 1.19, P value = 0.002)] tested. Additionally, with an FDR <0.05, all genotypic models demonstrated statistical significance. The association remained significant among subgroups of Asian and Caucasian populations, although analysis in some genetic models did not show a significant association. Begg's and Egger's tests did not show publication biases. In sensitivity analysis, one particular study was found to have an impact on the Recessive model's significance, but other models remained unaffected. The current meta-analysis found significant indications supporting the association between rs1137101 and obesity. To avail a deeper understanding of this association, future research should include large-scale studies conducted in diverse ethnic populations.
Collapse
Affiliation(s)
- Dilara Akhter Supti
- Department of Food Technology and Nutrition Science, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Farzana Akter
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Md Imranur Rahman
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Md Adnan Munim
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | | | - Rabia Jahan Tarin
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Sumaiya Afroz
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Hasan Al Reza
- Department of Genetic Engineering and Biotechnology, University of Dhaka, Dhaka, Bangladesh
| | - Roksana Yeasmin
- Department of Biochemistry, Ibrahim Medical College, Dhaka, Bangladesh
| | - Mohammad Rahanur Alam
- Department of Food Technology and Nutrition Science, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Md Shahadat Hossain
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| |
Collapse
|
27
|
Wallis NJ, Sumanasekera NT, Raffan E. Obesity risk factors in British Labrador retrievers: Effect of sex, neuter status, age, chocolate coat colour and food motivation. Vet Rec 2024; 194:e3410. [PMID: 37747436 DOI: 10.1002/vetr.3410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 08/11/2023] [Accepted: 08/17/2023] [Indexed: 09/26/2023]
Abstract
BACKGROUND Canine obesity is a complex disease affected by genetic, endocrine and environmental factors. It is associated with reduced lifespan and many comorbidities. Prevalence differs by breed, with Labrador retrievers at high risk. Past data on how biological risk factors impact weight gain have been contradictory, possibly because they were obtained from genetically heterogeneous populations. METHODS We investigated risk factors for canine obesity in a population of British Labrador retrievers (n = 521) with high-quality data on obesity, weight, owner-reported food motivation and related characteristics. We used linear regression to assess known and novel risk factors for obesity. RESULTS We found that neutering increased obesity in males (p < 0.001) but not females (p = 0.37). Older age was associated with obesity in female Labradors (p = 0.013) but not males (p = 0.49). We identified two new risk factors for obesity in Labrador retrievers: chocolate coat colour (p < 0.001) and high food motivation (p < 0.001). LIMITATIONS Strategic recruitment to collect both obese and lean dogs means this cohort is not suitable for assessing obesity prevalence in UK Labrador retrievers. CONCLUSION Studying this genetically homogeneous population informs our knowledge of common risk factors for obesity and expands those relevant to Labrador retrievers.
Collapse
Affiliation(s)
- Natalie J Wallis
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK
| | - Natasha T Sumanasekera
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK
| | - Eleanor Raffan
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK
| |
Collapse
|
28
|
Ferreira SRG, Macotela Y, Velloso LA, Mori MA. Determinants of obesity in Latin America. Nat Metab 2024; 6:409-432. [PMID: 38438626 DOI: 10.1038/s42255-024-00977-1] [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: 05/24/2023] [Accepted: 01/04/2024] [Indexed: 03/06/2024]
Abstract
Obesity rates are increasing almost everywhere in the world, although the pace and timing for this increase differ when populations from developed and developing countries are compared. The sharp and more recent increase in obesity rates in many Latin American countries is an example of that and results from regional characteristics that emerge from interactions between multiple factors. Aware of the complexity of enumerating these factors, we highlight eight main determinants (the physical environment, food exposure, economic and political interest, social inequity, limited access to scientific knowledge, culture, contextual behaviour and genetics) and discuss how they impact obesity rates in Latin American countries. We propose that initiatives aimed at understanding obesity and hampering obesity growth in Latin America should involve multidisciplinary, global approaches that consider these determinants to build more effective public policy and strategies, accounting for regional differences and disease complexity at the individual and systemic levels.
Collapse
Affiliation(s)
| | - Yazmín Macotela
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, UNAM Campus-Juriquilla, Querétaro, Mexico
| | - Licio A Velloso
- Obesity and Comorbidities Research Center, Faculty of Medical Sciences, Universidade Estadual de Campinas, Campinas, Brazil
| | - Marcelo A Mori
- Institute of Biology, Universidade Estadual de Campinas, Campinas, Brazil.
| |
Collapse
|
29
|
Di Fusco SA, Mocini E, Gulizia MM, Gabrielli D, Grimaldi M, Oliva F, Colivicchi F. ANMCO (Italian Association of Hospital Cardiologists) scientific statement: obesity in adults-an approach for cardiologists. Eat Weight Disord 2024; 29:1. [PMID: 38168872 PMCID: PMC10761446 DOI: 10.1007/s40519-023-01630-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/27/2023] [Indexed: 01/05/2024] Open
Abstract
Obesity is a complex, chronic disease requiring a multidisciplinary approach to its management. In clinical practice, body mass index and waist-related measurements can be used for obesity screening. The estimated prevalence of obesity among adults worldwide is 12%. With the expected further increase in overall obesity prevalence, clinicians will increasingly be managing patients with obesity. Energy balance is regulated by a complex neurohumoral system that involves the central nervous system and circulating mediators, among which leptin is the most studied. The functioning of these systems is influenced by both genetic and environmental factors. Obesity generally occurs when a genetically predisposed individual lives in an obesogenic environment for a long period. Cardiologists are deeply involved in evaluating patients with obesity. Cardiovascular risk profile is one of the most important items to be quantified to understand the health risk due to obesity and the clinical benefit that a single patient can obtain with weight loss. At the individual level, appropriate patient involvement, the detection of potential obesity causes, and a multidisciplinary approach are tools that can improve clinical outcomes. In the near future, we will probably have new pharmacological tools at our disposal that will facilitate achieving and maintaining weight loss. However, pharmacological treatment alone cannot cure such a complex disease. The aim of this paper is to summarize some key points of this field, such as obesity definition and measurement tools, its epidemiology, the main mechanisms underlying energy homeostasis, health consequences of obesity with a focus on cardiovascular diseases and the obesity paradox.Level of evidence V: report of expert committees.
Collapse
Affiliation(s)
- Stefania Angela Di Fusco
- Emergency Department, Clinical and Rehabilitation Cardiology Unit, San Filippo Neri Hospital, ASL Roma 1, Rome, Italy
| | - Edoardo Mocini
- Department of Experimental Medicine, Sapienza University, 00161, Rome, Italy.
| | | | - Domenico Gabrielli
- Cardio-Thoracic-Vascular Department, San Camillo-Forlanini Hospital, Rome, Italy
- Heart Care Foundation, Florence, Italy
| | - Massimo Grimaldi
- Department of Cardiology, General Regional Hospital "F. Miulli", Acquaviva delle Fonti, 70021, Bari, Italy
| | - Fabrizio Oliva
- De Gasperis Cardio Center, Niguarda Hospital, 20162, Milan, Italy
| | - Furio Colivicchi
- Emergency Department, Clinical and Rehabilitation Cardiology Unit, San Filippo Neri Hospital, ASL Roma 1, Rome, Italy
| |
Collapse
|
30
|
Monsalve FA, Delgado-López F, Fernández-Tapia B, González DR. Adipose Tissue, Non-Communicable Diseases, and Physical Exercise: An Imperfect Triangle. Int J Mol Sci 2023; 24:17168. [PMID: 38138997 PMCID: PMC10743187 DOI: 10.3390/ijms242417168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/20/2023] [Accepted: 09/26/2023] [Indexed: 12/24/2023] Open
Abstract
The study of adipose tissue has received considerable attention due to its importance not just in maintaining body energy homeostasis but also in playing a role in a number of other physiological processes. Beyond storing energy, adipose tissue is important in endocrine, immunological, and neuromodulatory functions, secreting hormones that participate in the regulation of energy homeostasis. An imbalance of these functions will generate structural and functional changes in the adipose tissue, favoring the secretion of deleterious adipocytokines that induce a pro-inflammatory state, allowing the development of metabolic and cardiovascular diseases and even some types of cancer. A common theme worldwide has been the development of professional guidelines for the control and treatment of obesity, with emphasis on hypocaloric diets and exercise. The aim of this review is to examine the pathophysiological mechanisms of obesity, considering the relationship among adipose tissue and two aspects that contribute positively or negatively to keeping a healthy body homeostasis, namely, exercise and noninfectious diseases. We conclude that the relationship of these aspects does not have homogeneous effects among individuals. Nevertheless, it is possible to establish some common mechanisms, like a decrease in pro-inflammatory markers in the case of exercise, and an increase in chronic inflammation in non-communicable diseases. An accurate diagnosis might consider the particular variables of a patient, namely their molecular profile and how it affects its metabolism, routines, and lifestyle; their underling health conditions; and probably even the constitution of their microbiome. We foresee that the development and accessibility of omics approaches and precision medicine will greatly improve the diagnosis, treatment, and successful outcomes for obese patients.
Collapse
Affiliation(s)
- Francisco A. Monsalve
- Department of Basic Biomedical Science, Faculty of Health Sciences, Universidad de Talca, Talca 3465548, Chile;
| | - Fernando Delgado-López
- Laboratories of Biomedical Research, Department of Preclinical Sciences, Faculty of Medicine, Universidad Católica del Maule, Talca 3466706, Chile;
| | | | - Daniel R. González
- Department of Basic Biomedical Science, Faculty of Health Sciences, Universidad de Talca, Talca 3465548, Chile;
| |
Collapse
|
31
|
Busebee B, Ghusn W, Cifuentes L, Acosta A. Obesity: A Review of Pathophysiology and Classification. Mayo Clin Proc 2023; 98:1842-1857. [PMID: 37831039 PMCID: PMC10843116 DOI: 10.1016/j.mayocp.2023.05.026] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 05/18/2023] [Accepted: 05/30/2023] [Indexed: 10/14/2023]
Abstract
Obesity is a chronic, multifactorial, and morbid disease. In the United States, 69% of adults are overweight or have obesity, and the global prevalence of obesity is increasing. Obesity is influenced by genetic, neurologic, metabolic, enteric, and behavioral processes. It remains a key modifiable risk factor for many comorbid diseases, including cardiovascular disease, diabetes mellitus, and cancer. Whereas there are recent and significant advances in obesity therapy, including diets, lifestyle modifications, pharmacotherapies, endoscopic procedures, and bariatric surgeries, there is an immense need for a better understanding of the heterogeneity in the pathophysiologic process of obesity and outcomes. Here we review salient pathophysiologic mechanisms underlying the development and morbidity of obesity as well as pathophysiologically based classification systems that inform current obesity management and may inform improved and individualized management in the future.
Collapse
Affiliation(s)
| | - Wissam Ghusn
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Lizeth Cifuentes
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Andres Acosta
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN.
| |
Collapse
|
32
|
Yu H, Yu H, Zhang R, Peng D, Yan D, Gu Y, Bao Y, Jia W, Zhang H, Hu C. Targeted gene panel provides advantages over whole-exome sequencing for diagnosing obesity and diabetes mellitus. J Mol Cell Biol 2023; 15:mjad040. [PMID: 37327085 PMCID: PMC10847719 DOI: 10.1093/jmcb/mjad040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/12/2023] [Accepted: 06/15/2023] [Indexed: 06/18/2023] Open
Abstract
A small fraction of patients diagnosed with obesity or diabetes mellitus has an underlying monogenic cause. Here, we constructed a targeted gene panel consisting of 83 genes reported to be causative for monogenic obesity or diabetes. We performed this panel in 481 patients to detect causative variants and compared these results with whole-exome sequencing (WES) data available for 146 of these patients. The coverage of targeted gene panel sequencing was significantly higher than that of WES. The diagnostic yield in patients sequenced by the panel was 32.9% with subsequent WES leading to three additional diagnoses with two novel genes. In total, 178 variants in 83 genes were detected in 146 patients by targeted sequencing. Three of the 178 variants were missed by WES, although the WES-only approach had a similar diagnostic yield. For the 335 samples only receiving targeted sequencing, the diagnostic yield was 32.2%. In conclusion, taking into account the lower costs, shorter turnaround time, and higher quality of data, targeted sequencing is a more effective screening method for monogenic obesity and diabetes compared to WES. Therefore, this approach could be routinely established and used as a first-tier test in clinical practice for specific patients.
Collapse
Affiliation(s)
- Hairong Yu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Haoyong Yu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Rong Zhang
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Danfeng Peng
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Dandan Yan
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Yunjuan Gu
- Department of Endocrinology, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Yuqian Bao
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Weiping Jia
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Hong Zhang
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Cheng Hu
- Shanghai Diabetes Institute, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Clinical Center for Diabetes, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
- Institute for Metabolic Disease, Fengxian Central Hospital Affiliated to Southern Medical University, Shanghai 201499, China
| |
Collapse
|
33
|
Agarwal T, Lyngdoh T, Khadgawat R, Prabhakaran D, Chandak GR, Walia GK. Genetic architecture of adiposity measures among Asians: Findings from GWAS. Ann Hum Genet 2023; 87:255-273. [PMID: 37671428 DOI: 10.1111/ahg.12526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 08/17/2023] [Accepted: 08/18/2023] [Indexed: 09/07/2023]
Abstract
Adiposity has gradually become a global public threat over the years with drastic increase in the attributable deaths and disability adjusted life years (DALYs). Given an increased metabolic risk among Asians as compared to Europeans for any given body mass index (BMI) and considering the differences in genetic architecture between them, the present review aims to summarize the findings from genome-wide scans for various adiposity indices and related anthropometric measures from Asian populations. The search for related studies, published till February 2022, were made on PubMed and GWAS Catalog using search strategy built with relevant keywords joined by Boolean operators. It was recorded that out of a total of 47 identified studies, maximum studies are from Korean population (n = 14), followed by Chinese (n = 7), and Japanese (n = 6). Nearly 200 loci have been identified for BMI, 660 for height, 16 for weight, 28 for circumferences (waist and hip), 32 for ratios (waist hip ratio [WHR] and thoracic hip ratio [THR]), 5 for body fat, 16 for obesity, and 28 for adiposity-related blood markers among Asians. It was observed that though, most of the loci were unique for each trait, there were 3 loci in common to BMI and WHR. Apart from validation of variants identified in European setting, there were many novel loci discovered in Asian populations. Notably, 125 novel loci form Asian studies have been reported for BMI, 47 for height, 5 for waist circumference, and 2 for adiponectin level to the existing knowledge of the genetic framework of adiposity and related measures. It is necessary to examine more advanced adiposity measures, specifically of relevance to abdominal adiposity, a major risk factor for cardiometabolic disorders among Asians. Moreover, in spite of being one continent, there is diversity among different ethnicities across Asia in terms of lifestyle, climate, geography, genetic structure and consequently the phenotypic manifestations. Hence, it is also important to consider ethnic specific studies for identifying and validating reliable genetic variants of adiposity measures among Asians.
Collapse
Affiliation(s)
- Tripti Agarwal
- Indian Institute of Public Health-Delhi, Public Health Foundation of India, Delhi, India
| | | | | | | | - Giriraj Ratan Chandak
- Genomic Research in Complex diseases (GRC Group), CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | | |
Collapse
|
34
|
Farooqi S. Obesity and thinness: insights from genetics. Philos Trans R Soc Lond B Biol Sci 2023; 378:20220205. [PMID: 37661743 PMCID: PMC10475868 DOI: 10.1098/rstb.2022.0205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 06/09/2023] [Indexed: 09/05/2023] Open
Abstract
Genetic disruption of key molecular components of the hypothalamic leptin-melanocortin pathway causes severe obesity in mice and humans. Physiological studies in people who carry these mutations have shown that the adipose tissue-derived hormone leptin primarily acts to defend against starvation. A lack of leptin causes an intense drive to eat and increases the rewarding properties of food, demonstrating that human appetite has a strong biological basis. Genetic studies in clinical- and population-based cohorts of people with obesity or thinness continue to provide new insights into the physiological mechanisms involved in weight regulation and identify molecular targets for weight loss therapy. This article is part of a discussion meeting issue 'Causes of obesity: theories, conjectures and evidence (Part II)'.
Collapse
Affiliation(s)
- Sadaf Farooqi
- Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Box 289, Cambridge CB2 0QQ, UK
| |
Collapse
|
35
|
Ahrendsen JT, Nong Y, Huo Y, Steele J, Anderson MP. CD8 cytotoxic T-cell infiltrates and cellular damage in the hypothalamus in human obesity. Acta Neuropathol Commun 2023; 11:163. [PMID: 37814324 PMCID: PMC10563257 DOI: 10.1186/s40478-023-01659-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Accepted: 09/24/2023] [Indexed: 10/11/2023] Open
Abstract
Rare cases of paraneoplastic obesity in children suggest sporadic obesity might also arise from an adaptive immune cell-mediated mechanism. Since the hypothalamus is a central regulator of feeding behavior and energy expenditure, we quantified lymphocytic inflammation in this region in a cohort of obese and non-obese human post-mortem brains. We report that CD8-positive cytotoxic T-cells are increased in hypothalamic median eminence/arcuate nucleus (ME/Arc) and bed nucleus of the stria terminalis in 40% of obese compared to non-obese patients, but not in other hypothalamic nuclei or brain regions. CD8 T-cells were most abundant in individuals with concurrent obesity and diabetes. Markers of cytotoxic T-cell induced damage, activated caspase 3 and poly-ADP ribose, were also elevated in the ME/Arc of obese patients. To provoke CD8 cytotoxic T-cell infiltrates in ventromedial region of hypothalamus in mice we performed stereotactic injections of an adeno-associated virus expressing immunogenic green fluorescent protein or saline. AAV but not saline injections triggered hypothalamic CD8 T-cell infiltrates associated with a rapid weight gain in mice recapitulating the findings in human obesity. This is the first description of the neuropathology of human obesity and when combined with its reconstitution in a mouse model suggests adaptive immunity may drive as much as 40% of the human condition.
Collapse
Affiliation(s)
- Jared T Ahrendsen
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yi Nong
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
- Neuroscience Therapeutic Focus Area, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Yuda Huo
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
- Neuroscience Therapeutic Focus Area, Regeneron Pharmaceuticals, Tarrytown, NY, USA
| | - Jasmine Steele
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA
| | - Matthew P Anderson
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA.
- Department of Neurology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts, USA.
- Neuroscience Therapeutic Focus Area, Regeneron Pharmaceuticals, Tarrytown, NY, USA.
| |
Collapse
|
36
|
Zandvakili I, Pulaski M, Pickett-Blakely O. A phenotypic approach to obesity treatment. Nutr Clin Pract 2023; 38:959-975. [PMID: 37277855 DOI: 10.1002/ncp.11013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 03/23/2023] [Accepted: 04/16/2023] [Indexed: 06/07/2023] Open
Abstract
Obesity is a chronic disease that increases morbidity and mortality and adversely affects quality of life. The rapid rise of obesity has outpaced the development and deployment of effective therapeutic interventions, thereby creating a global health crisis. The presentation, complications, and response to obesity treatments vary, yet lifestyle modification, which is the foundational therapeutic intervention for obesity, is often "one size fits all." The concept of personalized medicine uses genetic and phenotypic information as a guide for disease prevention, diagnosis, and treatment and has been successfully applied in diseases such as cancer, but not in obesity. As we gain insight into the pathophysiologic mechanisms of obesity and its phenotypic expression, specific pathways can be targeted to yield a greater, more sustained therapeutic impact in an individual patient with obesity. A phenotype-based pharmacologic treatment approach utilizing objective measures to classify patients into predominant obesity mechanism groups resulted in greater weight loss (compared with a non-phenotype-based approach) in a recent study by Acosta and colleagues. In this review, we discuss the application of lifestyle modifications, behavior therapy and pharmacotherapy using the obesity phenotype-based approach as a framework.
Collapse
Affiliation(s)
- Inuk Zandvakili
- Division of Digestive Diseases, Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, Ohio, USA
- Division of Gastroenterology and Hepatology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Marya Pulaski
- Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Octavia Pickett-Blakely
- Division of Gastroenterology and Hepatology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
37
|
Vasile CM, Padovani P, Rujinski SD, Nicolosu D, Toma C, Turcu AA, Cioboata R. The Increase in Childhood Obesity and Its Association with Hypertension during Pandemics. J Clin Med 2023; 12:5909. [PMID: 37762850 PMCID: PMC10531996 DOI: 10.3390/jcm12185909] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/07/2023] [Accepted: 09/09/2023] [Indexed: 09/29/2023] Open
Abstract
There has been a major ongoing health impact of the COVID-19 pandemic on children's lives, including lifestyle and overall health. Enforcement of prevention measures, such as school closures and social distancing, has significantly affected children's daily routines and activities. This perspective manuscript aims to explore the rise in childhood obesity and its association with hypertension during pandemics. The COVID-19 pandemic has led to significant disruptions in children's routines, including reduced physical activity, increased sedentary behavior, and changes in dietary patterns. These factors, coupled with the psychological impact of the pandemic, have contributed to an alarming increase in childhood obesity rates. This paper has highlighted the concerning increase in childhood obesity and hypertension during pandemics. The disruptions caused by the COVID-19 pandemic, including reduced physical activity, increased sedentary behaviors, and changes in dietary patterns, have contributed to the rise in these health conditions. It is crucial to recognize the long-term consequences of childhood obesity and hypertension and the urgent need for a comprehensive approach to address them.
Collapse
Affiliation(s)
- Corina Maria Vasile
- Department of Pediatric and Adult Congenital Cardiology, University Hospital of Bordeaux, F-33600 Bordeaux, France;
| | - Paul Padovani
- Nantes Université, CHU Nantes, Department of Pediatric Cardiology and Pediatric Cardiac Surgery, FHU PreciCare, F-44000 Nantes, France;
- Nantes Université, CHU Nantes, INSERM, CIC FEA 1413, F-44000 Nantes, France
| | | | - Dragos Nicolosu
- Pneumology Department, Victor Babes University Hospital Craiova, 200515 Craiova, Romania; (D.N.); (R.C.)
| | - Claudia Toma
- Pneumology Department, University of Medicine Carol Davila, 020021 Bucharest, Romania;
| | - Adina Andreea Turcu
- Faculty of Dentistry, University of Pharmacy and Medicine Craiova, 200349 Craiova, Romania
| | - Ramona Cioboata
- Pneumology Department, Victor Babes University Hospital Craiova, 200515 Craiova, Romania; (D.N.); (R.C.)
- Pneumology Department, University of Medicine and Pharmacy, 200349 Craiova, Romania
| |
Collapse
|
38
|
Hanssen R, Auwerx C, Jõeloo M, Sadler MC, Henning E, Keogh J, Bounds R, Smith M, Firth HV, Kutalik Z, Farooqi IS, Reymond A, Lawler K. Chromosomal deletions on 16p11.2 encompassing SH2B1 are associated with accelerated metabolic disease. Cell Rep Med 2023; 4:101155. [PMID: 37586323 PMCID: PMC10439272 DOI: 10.1016/j.xcrm.2023.101155] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/08/2023] [Accepted: 07/18/2023] [Indexed: 08/18/2023]
Abstract
New approaches are needed to treat people whose obesity and type 2 diabetes (T2D) are driven by specific mechanisms. We investigate a deletion on chromosome 16p11.2 (breakpoint 2-3 [BP2-3]) encompassing SH2B1, a mediator of leptin and insulin signaling. Phenome-wide association scans in the UK (N = 502,399) and Estonian (N = 208,360) biobanks show that deletion carriers have increased body mass index (BMI; p = 1.3 × 10-10) and increased rates of T2D. Compared with BMI-matched controls, deletion carriers have an earlier onset of T2D, with poorer glycemic control despite higher medication usage. Cystatin C, a biomarker of kidney function, is significantly elevated in deletion carriers, suggesting increased risk of renal impairment. In a Mendelian randomization study, decreased SH2B1 expression increases T2D risk (p = 8.1 × 10-6). We conclude that people with 16p11.2 BP2-3 deletions have early, complex obesity and T2D and may benefit from therapies that enhance leptin and insulin signaling.
Collapse
Affiliation(s)
- Ruth Hanssen
- University of Cambridge Metabolic Research Laboratories, Wellcome-MRC Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Chiara Auwerx
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland; Department of Computational Biology, University of Lausanne, 1015 Lausanne, Switzerland; Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland; University Center for Primary Care and Public Health, 1010 Lausanne, Switzerland
| | - Maarja Jõeloo
- Institute of Molecular and Cell Biology, University of Tartu, 51010 Tartu, Estonia; Estonian Genome Centre, Institute of Genomics, University of Tartu, 51010 Tartu, Estonia
| | - Marie C Sadler
- Department of Computational Biology, University of Lausanne, 1015 Lausanne, Switzerland; Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland; University Center for Primary Care and Public Health, 1010 Lausanne, Switzerland
| | - Elana Henning
- University of Cambridge Metabolic Research Laboratories, Wellcome-MRC Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Julia Keogh
- University of Cambridge Metabolic Research Laboratories, Wellcome-MRC Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Rebecca Bounds
- University of Cambridge Metabolic Research Laboratories, Wellcome-MRC Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Miriam Smith
- University of Cambridge Metabolic Research Laboratories, Wellcome-MRC Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Helen V Firth
- Department of Clinical Genetics, Cambridge University Hospitals NHS Foundation Trust & Wellcome Sanger Institute, Cambridge, UK
| | - Zoltán Kutalik
- Department of Computational Biology, University of Lausanne, 1015 Lausanne, Switzerland; Swiss Institute of Bioinformatics, 1015 Lausanne, Switzerland; University Center for Primary Care and Public Health, 1010 Lausanne, Switzerland
| | - I Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories, Wellcome-MRC Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK.
| | - Alexandre Reymond
- Center for Integrative Genomics, University of Lausanne, 1015 Lausanne, Switzerland.
| | - Katherine Lawler
- University of Cambridge Metabolic Research Laboratories, Wellcome-MRC Institute of Metabolic Science and NIHR Cambridge Biomedical Research Centre, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK.
| |
Collapse
|
39
|
Horwitz A, Birk R. Adipose Tissue Hyperplasia and Hypertrophy in Common and Syndromic Obesity-The Case of BBS Obesity. Nutrients 2023; 15:3445. [PMID: 37571382 PMCID: PMC10421039 DOI: 10.3390/nu15153445] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/16/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023] Open
Abstract
Obesity is a metabolic state generated by the expansion of adipose tissue. Adipose tissue expansion depends on the interplay between hyperplasia and hypertrophy, and is mainly regulated by a complex interaction between genetics and excess energy intake. However, the genetic regulation of adipose tissue expansion is yet to be fully understood. Obesity can be divided into common multifactorial/polygenic obesity and monogenic obesity, non-syndromic and syndromic. Several genes related to obesity were found through studies of monogenic non-syndromic obesity models. However, syndromic obesity, characterized by additional features other than obesity, suggesting a more global role of the mutant genes related to the syndrome and, thus, an additional peripheral influence on the development of obesity, were hardly studied to date in this regard. This review summarizes present knowledge regarding the hyperplasia and hypertrophy of adipocytes in common obesity. Additionally, we highlight the scarce research on syndromic obesity as a model for studying adipocyte hyperplasia and hypertrophy, focusing on Bardet-Biedl syndrome (BBS). BBS obesity involves central and peripheral mechanisms, with molecular and mechanistic alternation in adipocyte hyperplasia and hypertrophy. Thus, we argue that using syndromic obesity models, such as BBS, can further advance our knowledge regarding peripheral adipocyte regulation in obesity.
Collapse
Affiliation(s)
| | - Ruth Birk
- Department of Nutrition, Faculty of Health Sciences, Ariel University, Ariel 40700, Israel;
| |
Collapse
|
40
|
Zhang Z, Chen N, Yin N, Liu R, He Y, Li D, Tong M, Gao A, Lu P, Zhao Y, Li H, Zhang J, Zhang D, Gu W, Hong J, Wang W, Qi L, Ning G, Wang J. The rs1421085 variant within FTO promotes brown fat thermogenesis. Nat Metab 2023; 5:1337-1351. [PMID: 37460841 DOI: 10.1038/s42255-023-00847-2] [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: 09/25/2021] [Accepted: 06/14/2023] [Indexed: 08/06/2023]
Abstract
One lead genetic risk signal of obesity-the rs1421085 T>C variant within the FTO gene-is reported to be functional in vitro but lacks evidence at an organism level. Here we recapitulate the homologous human variant in mice with global and brown adipocyte-specific variant knock-in and reveal that mice carrying the C-allele show increased brown fat thermogenic capacity and resistance to high-fat diet-induced adiposity, whereas the obesity-related phenotypic changes are blunted at thermoneutrality. Both in vivo and in vitro data reveal that the C-allele in brown adipocytes enhances the transcription of the Fto gene, which is associated with stronger chromatin looping linking the enhancer region and Fto promoter. Moreover, FTO knockdown or inhibition effectively eliminates the increased thermogenic ability of brown adipocytes carrying the C-allele. Taken together, these findings identify rs1421085 T>C as a functional variant promoting brown fat thermogenesis.
Collapse
Affiliation(s)
- Zhiyin Zhang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Na Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Nan Yin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Ruixin Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Yang He
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Danjie Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Muye Tong
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Aibo Gao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Peng Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Yuxiao Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Huabing Li
- Shanghai Institute of Immunology, State Key Laboratory of Oncogenes and Related Genes, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junfang Zhang
- Laboratory of Aquacultural Resources and Utilization, Ministry of Education, College of Fishery and Life Science, Shanghai Ocean University, Shanghai, China
| | - Dan Zhang
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Weiqiong Gu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Jie Hong
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, USA
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China
| | - Jiqiu Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai, China.
| |
Collapse
|
41
|
Lechner L, Opitz R, Silver MJ, Krabusch PM, Prentice AM, Field MS, Stachelscheid H, Leitão E, Schröder C, Fernandez Vallone V, Horsthemke B, Jöckel KH, Schmidt B, Nöthen MM, Hoffmann P, Herms S, Kleyn PW, Megges M, Blume-Peytavi U, Weiss K, Mai K, Blankenstein O, Obermayer B, Wiegand S, Kühnen P. Early-set POMC methylation variability is accompanied by increased risk for obesity and is addressable by MC4R agonist treatment. Sci Transl Med 2023; 15:eadg1659. [PMID: 37467315 DOI: 10.1126/scitranslmed.adg1659] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 06/22/2023] [Indexed: 07/21/2023]
Abstract
Increasing evidence points toward epigenetic variants as a risk factor for developing obesity. We analyzed DNA methylation of the POMC (pro-opiomelanocortin) gene, which is pivotal for satiety regulation. We identified sex-specific and nongenetically determined POMC hypermethylation associated with a 1.4-fold (confidence interval, 1.03 to 2.04) increased individual risk of developing obesity. To investigate the early embryonic establishment of POMC methylation states, we established a human embryonic stem cell (hESC) model. Here, hESCs (WA01) were transferred into a naïve state, which was associated with a reduction of DNA methylation. Naïve hESCs were differentiated via a formative state into POMC-expressing hypothalamic neurons, which was accompanied by re-establishment of DNA methylation patterning. We observed that reduced POMC gene expression was associated with increased POMC methylation in POMC-expressing neurons. On the basis of these findings, we treated POMC-hypermethylated obese individuals (n = 5) with an MC4R agonist and observed a body weight reduction of 4.66 ± 2.16% (means ± SD) over a mean treatment duration of 38.4 ± 26.0 weeks. In summary, we identified an epigenetic obesity risk variant at the POMC gene fulfilling the criteria for a metastable epiallele established in early embryonic development that may be addressable by MC4R agonist treatment to reduce body weight.
Collapse
Affiliation(s)
- Lara Lechner
- Department of Pediatric Endocrinology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany
| | - Robert Opitz
- Institute for Experimental Pediatric Endocrinology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany
| | - Matt J Silver
- Medical Research Council Unit, Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Banjul, PO Box 273, Gambia
| | - Philipp M Krabusch
- Department of Pediatric Endocrinology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany
| | - Andrew M Prentice
- Medical Research Council Unit, Gambia at the London School of Hygiene and Tropical Medicine, Fajara, Banjul, PO Box 273, Gambia
| | - Martha S Field
- Division of Nutritional Sciences, Cornell University, Ithaca, NY 14850, USA
| | - Harald Stachelscheid
- Berlin Institute of Health, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, BIH Core Unit Stem Cells and Organoids, 13353 Berlin, Germany
| | - Elsa Leitão
- Institute of Human Genetics, University Hospital Essen, 45147 Essen, Germany
| | | | - Valeria Fernandez Vallone
- Berlin Institute of Health, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, BIH Core Unit Stem Cells and Organoids, 13353 Berlin, Germany
| | - Bernhard Horsthemke
- Institute of Human Genetics, University Hospital Essen, 45147 Essen, Germany
| | - Karl-Heinz Jöckel
- Institute of Medical Informatics, Biometry and Epidemiology, University Hospital Essen, 45147 Essen, Germany
| | - Börge Schmidt
- Institute of Medical Informatics, Biometry and Epidemiology, University Hospital Essen, 45147 Essen, Germany
| | - Markus M Nöthen
- Institute of Human Genetics, School of Medicine and University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Per Hoffmann
- Institute of Human Genetics, School of Medicine and University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | - Stefan Herms
- Institute of Human Genetics, School of Medicine and University Hospital Bonn, University of Bonn, 53127 Bonn, Germany
| | | | - Matthias Megges
- Department of Pediatric Endocrinology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany
| | - Ulrike Blume-Peytavi
- Clinical Research Center for Hair and Skin Science, Department of Dermatology and Venerology and Allergology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, and Humboldt-Universität zu Berlin, 10117 Berlin, Germany
| | - Katja Weiss
- Klinik für Angeborene Herzfehler - Kinderkardiologie, Deutsches Herzzentrum der Charité, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany
| | - Knut Mai
- Department of Endocrinology, Diabetes, and Nutrition and Charité Center for Cardiovascular Research, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
- German Center for Diabetes Research, 85764 München-Neuherberg, Germany
| | - Oliver Blankenstein
- Department of Pediatric Endocrinology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany
- Department Endocrinology and Metabolism, Labor Berlin-Charité Vivantes GmbH, 13353 Berlin, Germany
| | - Benedikt Obermayer
- Core Unit Bioinformatics (CUBI), Berlin Institute of Health/Charité- Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 10117 Berlin, Germany
| | - Susanna Wiegand
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Center for Social-Pediatric Care/Pediatric Endocrinology and Diabetology, 13353 Berlin, Germany
| | - Peter Kühnen
- Department of Pediatric Endocrinology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany
| |
Collapse
|
42
|
Carrasco-Luna J, Navarro-Solera M, Gombert M, Martín-Carbonell V, Carrasco-García Á, Del Castillo-Villaescusa C, García-Pérez MÁ, Codoñer-Franch P. Association of the rs17782313, rs17773430 and rs34114122 Polymorphisms of/near MC4R Gene with Obesity-Related Biomarkers in a Spanish Pediatric Cohort. CHILDREN (BASEL, SWITZERLAND) 2023; 10:1221. [PMID: 37508717 PMCID: PMC10378299 DOI: 10.3390/children10071221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 06/28/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023]
Abstract
Obesity is a multifactorial disease whose onset and development are shaped by the individual genetic background. The melanocortin 4 receptor gene (MC4R) is involved in the regulation of food intake and energy expenditure. Some of the single nucleotide polymorphisms (SNPs) of this gene are related to obesity and metabolic risk factors. The present study was undertaken to assess the relationship between three polymorphism SNPs, namely, rs17782313, rs17773430 and rs34114122, and obesity and metabolic risk factors. One hundred seventy-eight children with obesity aged between 7 and 16 years were studied to determine anthropometric variables and biochemical and inflammatory parameters. Our results highlight that metabolic risk factors, especially alterations in carbohydrate metabolism, were related to rs17782313. The presence of the minor C allele in the three variants (C-C-C) was significantly associated with anthropometric measures indicative of obesity, such as the body mass and fat mass indexes, and increased the values of insulinemia to 21.91 µIU/mL with respect to the wild type values. Our study suggests that the C-C-C haplotype of the SNPs rs17782313, rs17773430 and rs34114122 of the MC4R gene potentiates metabolic risk factors at early ages in children with obesity.
Collapse
Affiliation(s)
- Joaquín Carrasco-Luna
- Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, 46010 Valencia, Spain; (J.C.-L.); (M.N.-S.); (V.M.-C.); (Á.C.-G.)
- Department for Biotechnology, Faculty of Experimental Science, Catholic University of Valencia, 46001 Valencia, Spain
| | - María Navarro-Solera
- Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, 46010 Valencia, Spain; (J.C.-L.); (M.N.-S.); (V.M.-C.); (Á.C.-G.)
| | - Marie Gombert
- Biosciences Division, Center for Health Sciences, SRI International, Menlo Park, CA 94025, USA;
| | - Vanessa Martín-Carbonell
- Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, 46010 Valencia, Spain; (J.C.-L.); (M.N.-S.); (V.M.-C.); (Á.C.-G.)
| | - Álvaro Carrasco-García
- Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, 46010 Valencia, Spain; (J.C.-L.); (M.N.-S.); (V.M.-C.); (Á.C.-G.)
| | - Cristina Del Castillo-Villaescusa
- Department of Pediatrics, University Hospital Doctor Peset, Foundation of Promotion of Health, Biomedical Research in the Valencian Region (FISABIO), 46020 Valencia, Spain;
| | - Miguel Ángel García-Pérez
- Department of Genetics, Faculty of Biological Sciences, University of Valencia, INCLIVA, 46100 Valencia, Spain;
| | - Pilar Codoñer-Franch
- Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, 46010 Valencia, Spain; (J.C.-L.); (M.N.-S.); (V.M.-C.); (Á.C.-G.)
- Department of Pediatrics, University Hospital Doctor Peset, Foundation of Promotion of Health, Biomedical Research in the Valencian Region (FISABIO), 46020 Valencia, Spain;
| |
Collapse
|
43
|
de Kluiver H, Jansen R, Penninx BWJH, Giltay EJ, Schoevers RA, Milaneschi Y. Metabolomics signatures of depression: the role of symptom profiles. Transl Psychiatry 2023; 13:198. [PMID: 37301859 DOI: 10.1038/s41398-023-02484-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 05/04/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Depression shows a metabolomic signature overlapping with that of cardiometabolic conditions. Whether this signature is linked to specific depression profiles remains undetermined. Previous research suggested that metabolic alterations cluster more consistently with depressive symptoms of the atypical spectrum related to energy alterations, such as hyperphagia, weight gain, hypersomnia, fatigue and leaden paralysis. We characterized the metabolomic signature of an "atypical/energy-related" symptom (AES) profile and evaluated its specificity and consistency. Fifty-one metabolites measured using the Nightingale platform in 2876 participants from the Netherlands Study of Depression and Anxiety were analyzed. An 'AES profile' score was based on five items of the Inventory of Depressive Symptomatology (IDS) questionnaire. The AES profile was significantly associated with 31 metabolites including higher glycoprotein acetyls (β = 0.13, p = 1.35*10-12), isoleucine (β = 0.13, p = 1.45*10-10), very-low-density lipoproteins cholesterol (β = 0.11, p = 6.19*10-9) and saturated fatty acid levels (β = 0.09, p = 3.68*10-10), and lower high-density lipoproteins cholesterol (β = -0.07, p = 1.14*10-4). The metabolites were not significantly associated with a summary score of all other IDS items not included in the AES profile. Twenty-five AES-metabolites associations were internally replicated using data from the same subjects (N = 2015) collected at 6-year follow-up. We identified a specific metabolomic signature-commonly linked to cardiometabolic disorders-associated with a depression profile characterized by atypical, energy-related symptoms. The specific clustering of a metabolomic signature with a clinical profile identifies a more homogenous subgroup of depressed patients at higher cardiometabolic risk, and may represent a valuable target for interventions aiming at reducing depression's detrimental impact on health.
Collapse
Affiliation(s)
- Hilde de Kluiver
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health, Mental Health program, Amsterdam, The Netherlands
| | - Rick Jansen
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health, Mental Health program, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Mood, Anxiety, Psychosis, Sleep & Stress program, Amsterdam, The Netherlands
| | - Brenda W J H Penninx
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Public Health, Mental Health program, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Mood, Anxiety, Psychosis, Sleep & Stress program, Amsterdam, The Netherlands
| | - Erik J Giltay
- Department of Psychiatry, Leiden University Medical Center, Leiden, The Netherlands
| | - Robert A Schoevers
- University of Groningen, University Medical Center Groningen, Department of Psychiatry and Research School of Behavioural and Cognitive Neurosciences (BCN), Groningen, The Netherlands
| | - Yuri Milaneschi
- Department of Psychiatry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
- Amsterdam Public Health, Mental Health program, Amsterdam, The Netherlands.
- Amsterdam Neuroscience, Mood, Anxiety, Psychosis, Sleep & Stress program, Amsterdam, The Netherlands.
| |
Collapse
|
44
|
Merino J, Dashti HS, Levy DE, Del Rocío Sevilla-González M, Hivert MF, Porneala BC, Saxena R, Thorndike AN. Genetic predisposition to macronutrient preference and workplace food choices. Mol Psychiatry 2023; 28:2606-2611. [PMID: 37217678 DOI: 10.1038/s41380-023-02107-x] [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: 11/21/2022] [Revised: 04/27/2023] [Accepted: 05/03/2023] [Indexed: 05/24/2023]
Abstract
Prior research identified genetic variants influencing macronutrient preference, but whether genetic differences underlying nutrient preference affect long-term food choices is unknown. Here we examined the associations of polygenic scores for carbohydrate, fat, and protein preference with 12 months' workplace food purchases among 397 hospital employees from the ChooseWell 365 study. Food purchases were obtained retrospectively from the hospital's cafeteria sales data for the 12 months before participants were enrolled in the ChooseWell 365 study. Traffic light labels, visible to employees when making purchases, measured the quality of workplace purchases. During the 12-month study period, there were 215,692 cafeteria purchases. Each SD increase in the polygenic score for carbohydrate preference was associated with 2.3 additional purchases/month (95%CI, 0.2 to 4.3; p = 0.03) and a higher number of green-labeled purchases (β = 1.9, 95%CI, 0.5-3.3; p = 0.01). These associations were consistent in subgroup and sensitivity analyses accounting for additional sources of bias. There was no evidence of associations between fat and protein polygenic scores and cafeteria purchases. Findings from this study suggest that genetic differences in carbohydrate preference could influence long-term workplace food purchases and may inform follow-up experiments to enhance our understanding of the molecular mechanisms underlying food choice behavior.
Collapse
Affiliation(s)
- Jordi Merino
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| | - Hassan S Dashti
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Douglas E Levy
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Mongan Institute Health Policy Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Magdalena Del Rocío Sevilla-González
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, 100 Cambridge, Boston, MA, USA
| | - Marie-France Hivert
- Diabetes Unit, Massachusetts General Hospital, Boston, MA, USA
- Department of Population Medicine, Harvard Medical School, Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Bianca C Porneala
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Richa Saxena
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Anne N Thorndike
- Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
45
|
Nainu F, Frediansyah A, Mamada SS, Permana AD, Salampe M, Chandran D, Emran TB, Simal-Gandara J. Natural products targeting inflammation-related metabolic disorders: A comprehensive review. Heliyon 2023; 9:e16919. [PMID: 37346355 PMCID: PMC10279840 DOI: 10.1016/j.heliyon.2023.e16919] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 05/31/2023] [Accepted: 06/01/2023] [Indexed: 06/23/2023] Open
Abstract
Currently, the incidence of metabolic disorders is increasing, setting a challenge to global health. With major advancement in the diagnostic tools and clinical procedures, much has been known in the etiology of metabolic disorders and their corresponding pathophysiologies. In addition, the use of in vitro and in vivo experimental models prior to clinical studies has promoted numerous biomedical breakthroughs, including in the discovery and development of drug candidates to treat metabolic disorders. Indeed, chemicals isolated from natural products have been extensively studied as prospective drug candidates to manage diabetes, obesity, heart-related diseases, and cancer, partly due to their antioxidant and anti-inflammatory properties. Continuous efforts have been made in parallel to improve their bioactivity and bioavailability using selected drug delivery approaches. Here, we provide insights on recent progress in the role of inflammatory-mediated responses on the initiation of metabolic disorders, with particular reference to diabetes mellitus, obesity, heart-related diseases, and cancer. In addition, we discussed the prospective role of natural products in the management of diabetes, obesity, heart-related diseases, and cancers and provide lists of potential biological targets for high throughput screening in drug discovery and development. Lastly, we discussed findings observed in the preclinical and clinical studies prior to identifying suitable approaches on the phytochemical drug delivery systems that are potential to be used in the treatment of metabolic disorders.
Collapse
Affiliation(s)
- Firzan Nainu
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Tamalanrea, Makassar 90245, Indonesia
| | - Andri Frediansyah
- Research Center for Food Technology and Processing (PRTPP), National Research and Innovation Agency (BRIN), Yogyakarta 55861, Indonesia
| | - Sukamto S. Mamada
- Department of Pharmacy, Faculty of Pharmacy, Hasanuddin University, Tamalanrea, Makassar 90245, Indonesia
| | - Andi Dian Permana
- Department of Pharmaceutical Science and Technology, Faculty of Pharmacy, Hasanuddin University, Tamalanrea, Makassar 90245, Indonesia
| | | | - Deepak Chandran
- Department of Veterinary Sciences and Animal Husbandry, Amrita School of Agricultural Sciences, Amrita Vishwa Vidyapeetham University, Coimbatore 642109, India
| | - Talha Bin Emran
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School & Legorreta Cancer Center, Brown University, Providence, RI 02912, USA
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1207, Bangladesh
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| | - Jesus Simal-Gandara
- Universidade de Vigo, Nutrition and Bromatology Group, Analytical Chemistry and Food Science Department, Faculty of Science, E32004 Ourense, Spain
| |
Collapse
|
46
|
Mallardo M, D'Alleva M, Lazzer S, Giovanelli N, Graniero F, Billat V, Fiori F, Marinoni M, Parpinel M, Daniele A, Nigro E. Improvement of adiponectin in relation to physical performance and body composition in young obese males subjected to twenty-four weeks of training programs. Heliyon 2023; 9:e15790. [PMID: 37215851 PMCID: PMC10196512 DOI: 10.1016/j.heliyon.2023.e15790] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 04/21/2023] [Accepted: 04/21/2023] [Indexed: 05/24/2023] Open
Abstract
Obesity and related metabolic diseases represent a worldwide health problem. The main factor predisposing to obesity is an unhealthy lifestyle including the lack of physical activity. A pivotal role in the etio-pathogenesis of obesity is carried out by adipose tissue, an endocrine organ secreting several adipokines involved in numerous metabolic and inflammatory processes. Among these, of particular importance is adiponectin, an adipokine involved in the regulation of insulin sensibility and in anti-inflammatory processes. The aim of the study was to determine the effects of 24 weeks of two different training programs polarized (POL) and threshold training (THR) on body composition, physical capacities and adiponectin expression. Thirteen male obese subjects (BMI: 32.0 ± 3.0 kg m-2) followed 24 weeks of two different training programs, POL and THR, consisting of walking or running (or a combination of the two methods) in their normal living conditions. Before (T0) and after the end of the program (T1), the assessment of body composition was assessed by bioelectrical impedance and the concentration of salivary and serum adiponectin was analyzed by enzyme-linked immunosorbent assay and western blotting. Although the results obtained did not show significant differences between the two training programs, body mass and body mass index decreased by a mean of -4.46 ± 2.90 kg and 1.43 ± 0.92 kg m-2 (P < 0.05). Fat mass decreased by -4.47 ± 2.78 kg (P < 0.05). V'O2max increased by a mean of 0.20 ± 0.26 L min-1 (P < 0.05) Also, we observed an increase in saliva and in serum of adiponectin concentrations at T1 compared to T0 by 4.72 ± 3.52 μg mL-1 and 5.22 ± 4.74 ng mL-1 (P < 0.05) respectively. Finally, we found significant correlations between Δ serum adiponectin and Δ Hip (R = -0.686, P = 0.001) and between Δ salivary adiponectin and ΔWaist (R = -0.678, P = 0.011). Our results suggest that a 24 weeks training program, independently from intensity and volume, induces an amelioration of body composition and fitness performance. These improvements are associated with an increase in total and HMW adiponectin expression in both saliva and in serum.
Collapse
Affiliation(s)
- Marta Mallardo
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche, Farmaceutiche, Università della Campania “Luigi Vanvitelli”, via A. Vivaldi, 81100, Caserta, Italy
- CEINGE Biotecnologie Avanzate “Franco Salvatore” scarl, Via G. Salvatore 486, 80145, Napoli, Italy
| | - Mattia D'Alleva
- Department of Medicine, University of Udine, Udine, Italy
- School of Sport Sciences, University of Udine, Udine, Italy
| | - Stefano Lazzer
- Department of Medicine, University of Udine, Udine, Italy
- School of Sport Sciences, University of Udine, Udine, Italy
| | - Nicola Giovanelli
- Department of Medicine, University of Udine, Udine, Italy
- School of Sport Sciences, University of Udine, Udine, Italy
| | - Francesco Graniero
- Physical Exercise Prescription Center, Azienda Sanitaria Universitaria Friuli Centrale, Gemona del Friuli, Udine, Italy
| | - Véronique Billat
- Unité de Biologie Intégrative des Adaptations à l’Exercice, Université Paris-Saclay, Univ Evry, 91000, Evry-Courcouronnes, France
- BillaTraining SAS, 32 rue Paul Vaillant-Couturier, 94140, Alforville, France
| | - Federica Fiori
- Department of Medicine, University of Udine, Udine, Italy
| | | | - Maria Parpinel
- Department of Medicine, University of Udine, Udine, Italy
| | - Aurora Daniele
- CEINGE Biotecnologie Avanzate “Franco Salvatore” scarl, Via G. Salvatore 486, 80145, Napoli, Italy
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, via Pansini, Napoli, 80131, Italy
| | - Ersilia Nigro
- Dipartimento di Scienze e Tecnologie Ambientali, Biologiche, Farmaceutiche, Università della Campania “Luigi Vanvitelli”, via A. Vivaldi, 81100, Caserta, Italy
- CEINGE Biotecnologie Avanzate “Franco Salvatore” scarl, Via G. Salvatore 486, 80145, Napoli, Italy
| |
Collapse
|
47
|
Li Y, Zhu S, Du D, Li Q, Xie K, Chen L, Feng X, Wu X, Sun Z, Zhou J, Yang J, Shu G, Wang S, Gao P, Zhu C, Jiang Q, Wang L. TLR4 in POMC neurons regulates thermogenesis in a sex-dependent manner. J Lipid Res 2023; 64:100368. [PMID: 37028769 PMCID: PMC10205441 DOI: 10.1016/j.jlr.2023.100368] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 02/10/2023] [Accepted: 03/17/2023] [Indexed: 04/08/2023] Open
Abstract
The rising prevalence of obesity has become a worldwide health concern. Obesity usually occurs when there is an imbalance between energy intake and energy expenditure. However, energy expenditure consists of several components, including metabolism, physical activity, and thermogenesis. Toll-like receptor 4 (TLR4) is a transmembrane pattern recognition receptor, and it is abundantly expressed in the brain. Here, we showed that pro-opiomelanocortin (POMC)-specific deficiency of TLR4 directly modulates brown adipose tissue thermogenesis and lipid homeostasis in a sex-dependent manner. Deleting TLR4 in POMC neurons is sufficient to increase energy expenditure and thermogenesis resulting in reduced body weight in male mice. POMC neuron is a subpopulation of tyrosine hydroxylase neurons and projects into brown adipose tissue, which regulates the activity of sympathetic nervous system and contributes to thermogenesis in POMC-TLR4-KO male mice. By contrast, deleting TLR4 in POMC neurons decreases energy expenditure and increases body weight in female mice, which affects lipolysis of white adipose tissue (WAT). Mechanistically, TLR4 KO decreases the expression of the adipose triglyceride lipase and lipolytic enzyme hormone-sensitive lipase in WAT in female mice. Furthermore, the function of immune-related signaling pathway in WAT is inhibited because of obesity, which exacerbates the development of obesity reversely. Together, these results demonstrate that TLR4 in POMC neurons regulates thermogenesis and lipid balance in a sex-dependent manner.
Collapse
Affiliation(s)
- Yongxiang Li
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Shuqing Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Dan Du
- Department of Genetics and Endocrinology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangdong Provincial Clinical Research Center for Child Health, Guangzhou, China
| | - Qiyong Li
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Kailai Xie
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Lvshuang Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Xiajie Feng
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Xin Wu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Zhonghua Sun
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jingjing Zhou
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Jinping Yang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Gang Shu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Songbo Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Ping Gao
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Canjun Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China
| | - Qingyan Jiang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.
| | - Lina Wang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, South China Agricultural University, Guangzhou, Guangdong, China; National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, Guangdong, China.
| |
Collapse
|
48
|
Cifuentes L, Campos A, Sacoto D, Ghusn W, De la Rosa A, Feris F, McRae A, Bublitz JT, Hurtado MD, Olson J, Acosta A. Cardiovascular Risk and Diseases in Patients With and Without Leptin-Melanocortin Pathway Variants. Mayo Clin Proc 2023; 98:533-540. [PMID: 36549983 PMCID: PMC10079551 DOI: 10.1016/j.mayocp.2022.10.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 10/09/2022] [Accepted: 10/31/2022] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To study differences in cardiovascular risk factors and diseases between patients with and without genetic variants in the leptin-melanocortin pathway. METHODS A cross-sectional study of patients with a history of severe obesity genotyped in June 2019 as participants of the Mayo Clinic Biobank was conducted in March 2022 to assess differences in cardiovascular risk and diseases between carriers of a heterozygous variant in the leptin-melanocortin pathway and noncarriers. Cardiovascular risk factors included hypertension, diabetes, dyslipidemia, and smoking. Cardiovascular disease includes coronary artery disease, peripheral artery disease, and cerebrovascular accidents. Patients with a history of bariatric surgery were excluded. We used logistic regression models to estimate the odds ratio and 95% CI, adjusting for age, body mass index (BMI), and sex. RESULTS Among a total of 168 carriers (8%; 121 [72%] female; mean [SD] age, 65.1 [14.9] years; BMI, 44.0 [7.4] kg/m2) and 2039 noncarriers (92%; 1446 [71%] female; mean [SD] age, 64.9 [14.4] years; BMI, 42.9 [6.6] kg/m2), carriers had higher prevalence odds of hypertension (odds ratio, 3.26; 95% CI, 2.31 to 4.61; P<.001) and reported higher number of cardiovascular risk factors compared with noncarriers (2.4 [1.1] vs 2.0 [1.1]; P<.001). There were no significant differences in the adjusted odds associated with diabetes, dyslipidemia, smoking, or cardiovascular disease. CONCLUSION Despite having similar body weight and BMI, carriers of heterozygous variants in the leptin-melanocortin pathway had higher rates of hypertension than noncarriers. These findings point to an association between hypertension and leptin-melanocortin pathway variants.
Collapse
Affiliation(s)
- Lizeth Cifuentes
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Alejandro Campos
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Daniel Sacoto
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Wissam Ghusn
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Alan De la Rosa
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Fauzi Feris
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Alison McRae
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN
| | - Joshua T Bublitz
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Maria D Hurtado
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN; Division of Endocrinology, Diabetes, Metabolism, and Nutrition, Department of Medicine, Mayo Clinic Health System, La Crosse, WI
| | - Janet Olson
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Andres Acosta
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN.
| |
Collapse
|
49
|
Vourdoumpa A, Paltoglou G, Charmandari E. The Genetic Basis of Childhood Obesity: A Systematic Review. Nutrients 2023; 15:1416. [PMID: 36986146 PMCID: PMC10058966 DOI: 10.3390/nu15061416] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/05/2023] [Accepted: 03/10/2023] [Indexed: 03/17/2023] Open
Abstract
Overweight and obesity in childhood and adolescence represents one of the most challenging public health problems of our century owing to its epidemic proportions and the associated significant morbidity, mortality, and increase in public health costs. The pathogenesis of polygenic obesity is multifactorial and is due to the interaction among genetic, epigenetic, and environmental factors. More than 1100 independent genetic loci associated with obesity traits have been currently identified, and there is great interest in the decoding of their biological functions and the gene-environment interaction. The present study aimed to systematically review the scientific evidence and to explore the relation of single-nucleotide polymorphisms (SNPs) and copy number variants (CNVs) with changes in body mass index (BMI) and other measures of body composition in children and adolescents with obesity, as well as their response to lifestyle interventions. Twenty-seven studies were included in the qualitative synthesis, which consisted of 7928 overweight/obese children and adolescents at different stages of pubertal development who underwent multidisciplinary management. The effect of polymorphisms in 92 different genes was assessed and revealed SNPs in 24 genetic loci significantly associated with BMI and/or body composition change, which contribute to the complex metabolic imbalance of obesity, including the regulation of appetite and energy balance, the homeostasis of glucose, lipid, and adipose tissue, as well as their interactions. The decoding of the genetic and molecular/cellular pathophysiology of obesity and the gene-environment interactions, alongside with the individual genotype, will enable us to design targeted and personalized preventive and management interventions for obesity early in life.
Collapse
Affiliation(s)
- Aikaterini Vourdoumpa
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece
| | - George Paltoglou
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece
| | - Evangelia Charmandari
- Division of Endocrinology, Metabolism and Diabetes, First Department of Pediatrics, National and Kapodistrian University of Athens Medical School, ‘Aghia Sophia’ Children’s Hospital, 11527 Athens, Greece
- Division of Endocrinology and Metabolism, Center for Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| |
Collapse
|
50
|
Talbot F, Feetham CH, Mokrosiński J, Lawler K, Keogh JM, Henning E, Mendes de Oliveira E, Ayinampudi V, Saeed S, Bonnefond A, Arslan M, Yeo GSH, Froguel P, Bechtold DA, Adamson A, Humphreys N, Barroso I, Luckman SM, Farooqi IS. A rare human variant that disrupts GPR10 signalling causes weight gain in mice. Nat Commun 2023; 14:1450. [PMID: 36922513 PMCID: PMC10017677 DOI: 10.1038/s41467-023-36966-3] [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] [Received: 08/11/2020] [Accepted: 02/23/2023] [Indexed: 03/17/2023] Open
Abstract
Disruption of brain-expressed G protein-coupled receptor-10 (GPR10) causes obesity in animals. Here, we identify multiple rare variants in GPR10 in people with severe obesity and in normal weight controls. These variants impair ligand binding and G protein-dependent signalling in cells. Transgenic mice harbouring a loss of function GPR10 variant found in an individual with obesity, gain excessive weight due to decreased energy expenditure rather than increased food intake. This evidence supports a role for GPR10 in human energy homeostasis. Therapeutic targeting of GPR10 may represent an effective weight-loss strategy.
Collapse
Affiliation(s)
- Fleur Talbot
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Claire H Feetham
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Jacek Mokrosiński
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Katherine Lawler
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Julia M Keogh
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Elana Henning
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Edson Mendes de Oliveira
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Vikram Ayinampudi
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Sadia Saeed
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, 59000, France
- Université de Lille, Lille, 59000, France
| | - Amélie Bonnefond
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, 59000, France
- Université de Lille, Lille, 59000, France
| | - Mohammed Arslan
- School of Life Sciences, Forman Christian College, Lahore, Pakistan
| | - Giles S H Yeo
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
- MRC Metabolic Diseases Unit, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
| | - Philippe Froguel
- Department of Metabolism, Digestion and Reproduction, Imperial College London, London, UK
- Inserm UMR1283, CNRS UMR8199, European Genomic Institute for Diabetes (EGID), Institut Pasteur de Lille, Lille University Hospital, Lille, 59000, France
- Université de Lille, Lille, 59000, France
| | - David A Bechtold
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Antony Adamson
- Genome Editing Unit, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Neil Humphreys
- Genome Editing Unit, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Inês Barroso
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK
- MRC Epidemiology Unit, University of Cambridge, Cambridge, UK
- Exeter Centre of Excellence for Diabetes Research (ExCEED), University of Exeter Medical School, Exeter, UK
| | - Simon M Luckman
- Division of Diabetes, Endocrinology and Gastroenterology, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| | - I Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, CB2 0QQ, UK.
| |
Collapse
|