1
|
Baldini V, Venezia N, Iriti A, Quattrocchi S, Zenesini C, Biscarini F, Atti AR, Menchetti M, Franceschini C, Varallo G, De Ronchi D, Plazzi G, Pizza F. Eating disorders in narcolepsy type 1: Evidence from a cross-sectional Italian study. J Sleep Res 2024; 33:e14150. [PMID: 38351712 DOI: 10.1111/jsr.14150] [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: 11/22/2023] [Revised: 12/22/2023] [Accepted: 01/05/2024] [Indexed: 10/18/2024]
Abstract
Narcolepsy type 1 is a chronic central disorder of hypersomnolence, and it is frequently accompanied by overweight, but the association between narcolepsy type 1 and eating disorders is controversial. Our study aims to compare patients with narcolepsy type 1 and controls on the symptomatology of eating disorders and to evaluate the association between clinical factors. This is a cross-sectional study, with consecutive recruitment of patients with narcolepsy type 1 attending the Outpatient Clinic for Narcolepsy at the IRCCS Istituto delle Scienze Neurologiche di Bologna (Italy) for routine follow-up visits. Healthy subjects from general populations were recruited as controls. Patients underwent a questionnaire-based assessment using the Eating Disorder Examination Questionnaire (EDE-Q), Binge Eating Scale (BES), Italian Night Eating Questionnaire (I-NEQ), Epworth Sleepiness Scale (ESS), and Narcolepsy Severity Scale (NSS). One hundred and thirty-eight patients with narcolepsy type 1 and 162 controls were enrolled. This study showed that individuals with narcolepsy type 1 reported higher scores on the EDE-Q, I-NEQ, and a higher body mass index (BMI) than the controls. The logistic regression analysis results, with EDE-Q positivity as a dependent variable, demonstrate a significant association with antidepressant drugs, female sex, and the use of sodium oxybate. We found an association between antidepressant drug consumption, the NSS total score, and female sex with BES positivity as the dependent variable. The logistic regression analysis for I-NEQ positivity found an association with antidepressant drug use. This study shows that patients with narcolepsy type 1 frequently present with comorbid eating disorder symptomatology, mainly night eating syndrome. Investigating the possible presence of eating disorders symptomatology through questionnaires is fundamental during the assessment of patients with narcolepsy type 1.
Collapse
Affiliation(s)
- Valentina Baldini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Noemi Venezia
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Anna Iriti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Silvia Quattrocchi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Corrado Zenesini
- IRCCS Istituto Delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
| | - Francesco Biscarini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Anna Rita Atti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Marco Menchetti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | | | - Giorgia Varallo
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Diana De Ronchi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Giuseppe Plazzi
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
- IRCCS Istituto Delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
| | - Fabio Pizza
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- IRCCS Istituto Delle Scienze Neurologiche di Bologna (ISNB), Bologna, Italy
| |
Collapse
|
2
|
Renard E, Thevenard-Berger A, Meyre D. Medical semiology of patients with monogenic obesity: A systematic review. Obes Rev 2024; 25:e13797. [PMID: 38956946 DOI: 10.1111/obr.13797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 03/20/2024] [Accepted: 06/19/2024] [Indexed: 07/04/2024]
Abstract
Patients with monogenic obesity display numerous medical features on top of hyperphagic obesity, but no study to date has provided an exhaustive description of their semiology. Two reviewers independently conducted a systematic review of MEDLINE, Embase, and Web of Science Core Collection databases from inception to January 2022 to identify studies that described symptoms of patients carrying pathogenic mutations in at least one of eight monogenic obesity genes (ADCY3, LEP, LEPR, MC3R, MC4R, MRAP2, PCSK1, and POMC). Of 5207 identified references, 269 were deemed eligible after title and abstract screening, full-text reading, and risk of bias and quality assessment. Data extraction included mutation spectrum and mode of inheritance, clinical presentation (e.g., anthropometry, energy intake and eating behaviors, digestive function, puberty and fertility, cognitive features, infectious diseases, morphological characteristics, chronic respiratory disease, and cardiovascular disease), biological characteristics (metabolic profile, endocrinology, hematology), radiological features, and treatments. The review provides an exhaustive description of mandatory, non-mandatory, and unique symptoms in heterozygous and homozygous carriers of mutation in eight monogenic obesity genes. This information is critical to help clinicians to orient genetic testing in subsets of patients with suspected monogenic obesity and provide actionable treatments (e.g., recombinant leptin and MC4R agonist).
Collapse
Affiliation(s)
- Emeline Renard
- INSERM UMR_S 1256, Nutrition, Genetics, and Environmental Risk Exposure (NGERE), Faculty of Medicine of Nancy, University of Lorraine, Nancy, France
- Department of Pediatrics, University Hospital of Nancy, Nancy, France
| | | | - David Meyre
- INSERM UMR_S 1256, Nutrition, Genetics, and Environmental Risk Exposure (NGERE), Faculty of Medicine of Nancy, University of Lorraine, Nancy, France
- Department of Molecular Medicine, Division of Biochemistry, Molecular Biology, and Nutrition, University Hospital of Nancy, Nancy, France
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Canada
| |
Collapse
|
3
|
Thompson MD, Percy ME, Cole DEC, Bichet DG, Hauser AS, Gorvin CM. G protein-coupled receptor (GPCR) gene variants and human genetic disease. Crit Rev Clin Lab Sci 2024; 61:317-346. [PMID: 38497103 DOI: 10.1080/10408363.2023.2286606] [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: 05/24/2023] [Revised: 08/28/2023] [Accepted: 11/19/2023] [Indexed: 03/19/2024]
Abstract
Genetic variations in the genes encoding G protein-coupled receptors (GPCRs) can disrupt receptor structure and function, which can result in human genetic diseases. Disease-causing mutations have been reported in at least 55 GPCRs for more than 66 monogenic diseases in humans. The spectrum of pathogenic and likely pathogenic variants includes loss of function variants that decrease receptor signaling on one extreme and gain of function that may result in biased signaling or constitutive activity, originally modeled on prototypical rhodopsin GPCR variants identified in retinitis pigmentosa, on the other. GPCR variants disrupt ligand binding, G protein coupling, accessory protein function, receptor desensitization and receptor recycling. Next generation sequencing has made it possible to identify variants of uncertain significance (VUS). We discuss variants in receptors known to result in disease and in silico strategies for disambiguation of VUS such as sorting intolerant from tolerant and polymorphism phenotyping. Modeling of variants has contributed to drug development and precision medicine, including drugs that target the melanocortin receptor in obesity and interventions that reverse loss of gonadotropin-releasing hormone receptor from the cell surface in idiopathic hypogonadotropic hypogonadism. Activating and inactivating variants of the calcium sensing receptor (CaSR) gene that are pathogenic in familial hypocalciuric hypercalcemia and autosomal dominant hypocalcemia have enabled the development of calcimimetics and calcilytics. Next generation sequencing has continued to identify variants in GPCR genes, including orphan receptors, that contribute to human phenotypes and may have therapeutic potential. Variants of the CaSR gene, some encoding an arginine-rich region that promotes receptor phosphorylation and intracellular retention, have been linked to an idiopathic epilepsy syndrome. Agnostic strategies have identified variants of the pyroglutamylated RF amide peptide receptor gene in intellectual disability and G protein-coupled receptor 39 identified in psoriatic arthropathy. Coding variants of the G protein-coupled receptor L1 (GPR37L1) orphan receptor gene have been identified in a rare familial progressive myoclonus epilepsy. The study of the role of GPCR variants in monogenic, Mendelian phenotypes has provided the basis of modeling the significance of more common variants of pharmacogenetic significance.
Collapse
Affiliation(s)
- Miles D Thompson
- Krembil Brain Institute, Toronto Western Hospital, Toronto, ON, Canada
| | - Maire E Percy
- Departments of Physiology and Obstetrics & Gynaecology, University of Toronto, Toronto, ON, Canada
| | - David E C Cole
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Daniel G Bichet
- Department of Physiology and Medicine, Hôpital du Sacré-Coeur, Université de Montréal, QC, Canada
| | - Alexander S Hauser
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Caroline M Gorvin
- Institute of Metabolism and Systems Research (IMSR), University of Birmingham, Birmingham, West Midlands, UK
| |
Collapse
|
4
|
Gravina AG, Panarese I, Trotta MC, D'Amico M, Pellegrino R, Ferraraccio F, Galdiero M, Alfano R, Grieco P, Federico A. Melanocortin 3,5 receptors immunohistochemical expression in colonic mucosa of inflammatory bowel disease patients: A matter of disease activity? World J Gastroenterol 2024; 30:1132-1142. [PMID: 38577176 PMCID: PMC10989484 DOI: 10.3748/wjg.v30.i9.1132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/15/2024] [Accepted: 02/08/2024] [Indexed: 03/06/2024] Open
Abstract
BACKGROUND Melanocortin 3 and 5 receptors (i.e., MC3R and MC5R) belong to the melanocortin family. However, data regarding their role in inflammatory bowel diseases (IBD) are currently unavailable. AIM This study aims to ascertain their expression profiles in the colonic mucosa of Crohn's disease (CD) and ulcerative colitis (UC), aligning them with IBD disease endoscopic and histologic activity. METHODS Colonic mucosal biopsies from CD/UC patients were sampled, and immunohistochemical analyses were conducted to evaluate the expression of MC3R and MC5R. Colonic sampling was performed on both traits with endoscopic scores (Mayo endoscopic score and CD endoscopic index of severity) consistent with inflamed mucosa and not consistent with disease activity (i.e., normal appearing mucosa). RESULTS In both CD and UC inflamed mucosa, MC3R (CD: + 7.7 fold vs normal mucosa, P < 0.01; UC: + 12 fold vs normal mucosa, P < 0.01) and MC5R (CD: + 5.5 fold vs normal mucosa, P < 0.01; UC: + 8.1 fold vs normal mucosa, P < 0.01) were significantly more expressed compared to normal mucosa. CONCLUSION MC3R and MC5R are expressed in the colon of IBD patients. Furthermore, expression may differ according to disease endoscopic activity, with a higher degree of expression in the traits affected by disease activity in both CD and UC, suggesting a potential use of these receptors in IBD pharmacology.
Collapse
Affiliation(s)
- Antonietta Gerarda Gravina
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Iacopo Panarese
- Pathology Division, Department of Mental and Physical Health and Preventive Medicine, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Maria Consiglia Trotta
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Michele D'Amico
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Raffaele Pellegrino
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Franca Ferraraccio
- Pathology Division, Department of Mental and Physical Health and Preventive Medicine, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Marilena Galdiero
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Roberto Alfano
- Department of Advanced Medical and Surgical Sciences (DAMSS), University of Campania Luigi Vanvitelli, Naples 80138, Italy
| | - Paolo Grieco
- Department of Pharmacy, University of Naples Federico II, Naples 80131, Italy
| | - Alessandro Federico
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples 80138, Italy
| |
Collapse
|
5
|
Mori K, Okuma H, Nakamura S, Uchinuma H, Kaga S, Nakajima H, Ogawa Y, Tsuchiya K. Melanocortin-4 receptor in macrophages attenuated angiotensin II-induced abdominal aortic aneurysm in mice. Sci Rep 2023; 13:19768. [PMID: 37957201 PMCID: PMC10643430 DOI: 10.1038/s41598-023-46831-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 11/06/2023] [Indexed: 11/15/2023] Open
Abstract
Obesity is recognized as an independent risk factor for abdominal aortic aneurysm (AAA). While mutations in the melanocortin-4 receptor (MC4R) gene is the most common cause of obesity caused by mutations in a single gene, the link between MC4R function and vascular disease has still remained unclear. Here, by using melanocortin-4 receptor (MC4R) deficient mice, we confirmed MC4R deficiency promotes AAA and atherosclerosis. We demonstrated the contribution of two novel factors towards vascular vulnerability in this model: leptin signaling in vascular smooth muscle cells (VSMCs) and loss of MC4R signaling in macrophages. Leptin was shown to promote vascular vulnerability via PI3K-dependent upregulation of Spp1 expression in VSMC. Additionally, Ang II-induced AAA incidence was significantly reduced when MC4R gene expression was myeloid cell-specifically rescued in MC4R deficient (MC4RTB/TB) mice. Ex vivo analysis showed a suppression in NF-κB activity in bone marrow-derived macrophages from LysM(+);MC4RTB/TB mice compared to LysM(-);MC4RTB/TB mice, which exaggerates with endogenous MC4R ligand treatment; α-MSH. These results suggest that MC4R signaling in macrophages attenuates AAA by inhibiting NF-κB activity and subsequent vascular inflammation.
Collapse
Affiliation(s)
- Kentaro Mori
- Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 4093898, Japan.
| | - Hideyuki Okuma
- Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 4093898, Japan
| | - Suguru Nakamura
- Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 4093898, Japan
| | - Hiroyuki Uchinuma
- Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 4093898, Japan
| | - Shigeaki Kaga
- Department of Surgery II, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Hiroyuki Nakajima
- Department of Surgery II, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kyoichiro Tsuchiya
- Interdisciplinary Graduate School of Medicine and Engineering, University of Yamanashi, 1110 Shimokato, Chuo, Yamanashi, 4093898, Japan.
| |
Collapse
|
6
|
Zhang M, Thieux M, Arvis L, Lin JS, Guyon A, Plancoulaine S, Villanueva C, Franco P. Metabolic disturbances in children with narcolepsy: a retrospective study. Sleep 2023; 46:zsad076. [PMID: 36971181 DOI: 10.1093/sleep/zsad076] [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: 05/10/2022] [Revised: 09/22/2022] [Indexed: 07/20/2023] Open
Abstract
STUDY OBJECTIVES To determine the prevalence of metabolic syndrome (MS) in children with narcolepsy and to evaluate their clinical and sleep characteristics according to the different components of MS. METHODS This retrospective study consisted of 58 de novo children with narcolepsy (median age: 12.7 years, 48.3% of boys). The recently published MS criteria in a French population of children were used. Clinical and sleep characteristics were compared between groups with different components of MS. RESULTS MS was present in 17.2% of children with narcolepsy, among whom 79.3% presented with high homeostasis model assessment for insulin resistance (HOMA-IR), 25.9% with high body mass index, 24.1% with low high-density lipoprotein cholesterol (HDL-C), and 12.1% with high triglycerides. Patients with at least two MS components had more night eating behaviors and tended to have lower percentage of slow-wave sleep and more fragmented sleep. On multiple sleep latency test, they had shorter mean sleep latencies to rapid eye movement (REM), non-REM sleep and tended to have more sleep onset REM periods (SOREMPs) than those with less than two MS components. CONCLUSIONS Insulin resistance was found to be the core metabolic disturbance in obese as well as in nonobese children with narcolepsy. Children with narcolepsy with at least two MS components presented a more severe daytime sleepiness and a higher prevalence of night-eating behaviors than those with less than two MS components. Such children might benefit from early evaluation and management in order to prevent future complications.
Collapse
Affiliation(s)
- Min Zhang
- Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM U1028, CNRS UMR5292, University of Lyon 1, Lyon, France
| | - Marine Thieux
- Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM U1028, CNRS UMR5292, University of Lyon 1, Lyon, France
- Pediatric Sleep Unit, Hôpital Femme Mère Enfant, Hospices Civils de Lyon and National Reference Center for Narcolepsy, University of Lyon1, Lyon, France
| | - Laura Arvis
- Pediatric endocrinology Unit, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Jian-Sheng Lin
- Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM U1028, CNRS UMR5292, University of Lyon 1, Lyon, France
| | - Aurore Guyon
- Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM U1028, CNRS UMR5292, University of Lyon 1, Lyon, France
- Pediatric Sleep Unit, Hôpital Femme Mère Enfant, Hospices Civils de Lyon and National Reference Center for Narcolepsy, University of Lyon1, Lyon, France
| | | | - Carine Villanueva
- Pediatric endocrinology Unit, Hôpital Femme Mère Enfant, Hospices Civils de Lyon, Université Claude Bernard Lyon 1, Lyon, France
| | - Patricia Franco
- Integrative Physiology of the Brain Arousal Systems, CRNL, INSERM U1028, CNRS UMR5292, University of Lyon 1, Lyon, France
- Pediatric Sleep Unit, Hôpital Femme Mère Enfant, Hospices Civils de Lyon and National Reference Center for Narcolepsy, University of Lyon1, Lyon, France
| |
Collapse
|
7
|
Nwayyir HA, Mutasher EM, Alabid OM, Jabbar MA, Abdulraheem Al-Kawaz WH, Alidrisi HA, Alabbood M, Chabek M, AlZubaidi M, Al-Khazrajy LA, Abd Alhaleem IS, Al-Hilfi ADA, Ali FM, AlBayati A, Al Saffar HB, Khazaal FAK. Recommendations for the prevention and management of obesity in the Iraqi population. Postgrad Med 2023:1-15. [PMID: 36803631 DOI: 10.1080/00325481.2023.2172914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
Abstract
Obesity is a chronic metabolic disease that has become one of the leading causes of disability and death in the world, affecting not only adults but also children and adolescents. In Iraq, one third of the adult population is overweight and another third obese. Clinical diagnosis is accomplished by measuring body mass index (BMI) and waist circumference (a marker for intra-visceral fat and higher metabolic and cardiovascular disease risk). A complex interaction between behavioral, social (rapid urbanization), environmental and genetic factors underlies the etiology of the disease. Treatment options for obesity may include a multicomponent approach, involving dietary changes to reduce calorie intake, an increase in physical activity, behavioral modification, pharmacotherapy and bariatric surgery. The purpose for these recommendations is to develop a management plan and standards of care that are relevant to the Iraqi population and that can prevent/manage obesity and obesity-related complications, for the promotion of a healthy community.
Collapse
Affiliation(s)
- Hussein Ali Nwayyir
- University of Basra, College of Medicine, Department of Endocrinology, Faiha Specialized Diabetes, Endocrine and Metabolism Centre, Iraq
| | - Esraa Majid Mutasher
- Department of Pediatric Endocrinology, Children Welfare Teaching Hospital, Medical City Complex, Iraq
| | | | | | | | | | - Majid Alabbood
- Department of Endocrinology, Almawani Hospital, Basra, Iraq
| | - Muhammed Chabek
- Consultant Obstetrics and Gynecology, Private Practice, Iraq
| | - Munib AlZubaidi
- Department of paediatrics, University of Baghdad College of Medicine, Iraq
| | - Lujain Anwar Al-Khazrajy
- Department of Family medicine, Consultant Family Physician, Al-Kindy College of Medicine, University of Baghdad, Iraq
| | | | | | | | - Ali AlBayati
- Department of Endocrinology Consultant Endocrinology, Professor of medicine, Babylon medical college, Iraq
| | | | | |
Collapse
|
8
|
Wilting J, Becker J. The lymphatic vascular system: much more than just a sewer. Cell Biosci 2022; 12:157. [PMID: 36109802 PMCID: PMC9476376 DOI: 10.1186/s13578-022-00898-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/06/2022] [Indexed: 11/18/2022] Open
Abstract
Almost 400 years after the (re)discovery of the lymphatic vascular system (LVS) by Gaspare Aselli (Asellius G. De lactibus, sive lacteis venis, quarto vasorum mesaraicorum genere, novo invento Gasparis Asellii Cremo. Dissertatio. (MDCXXIIX), Milan; 1628.), structure, function, development and evolution of this so-called 'second' vascular system are still enigmatic. Interest in the LVS was low because it was (and is) hardly visible, and its diseases are not as life-threatening as those of the blood vascular system. It is not uncommon for patients with lymphedema to be told that yes, they can live with it. Usually, the functions of the LVS are discussed in terms of fluid homeostasis, uptake of chylomicrons from the gut, and immune cell circulation. However, the broad molecular equipment of lymphatic endothelial cells suggests that they possess many more functions, which are also reflected in the pathophysiology of the system. With some specific exceptions, lymphatics develop in all organs. Although basic structure and function are the same regardless their position in the body wall or the internal organs, there are important site-specific characteristics. We discuss common structure and function of lymphatics; and point to important functions for hyaluronan turn-over, salt balance, coagulation, extracellular matrix production, adipose tissue development and potential appetite regulation, and the influence of hypoxia on the regulation of these functions. Differences with respect to the embryonic origin and molecular equipment between somatic and splanchnic lymphatics are discussed with a side-view on the phylogeny of the LVS. The functions of the lymphatic vasculature are much broader than generally thought, and lymphatic research will have many interesting and surprising aspects to offer in the future.
Collapse
Affiliation(s)
- Jörg Wilting
- Department of Anatomy and Cell Biology, University Medical School Göttingen, Göttingen, Germany.
| | - Jürgen Becker
- Department of Anatomy and Cell Biology, University Medical School Göttingen, Göttingen, Germany
| |
Collapse
|
9
|
Fatima MT, Ahmed I, Fakhro KA, Akil ASA. Melanocortin-4 receptor complexity in energy homeostasis,obesity and drug development strategies. Diabetes Obes Metab 2022; 24:583-598. [PMID: 34882941 PMCID: PMC9302617 DOI: 10.1111/dom.14618] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/05/2021] [Accepted: 12/06/2021] [Indexed: 12/19/2022]
Abstract
The melanocortin-4 receptor (MC4R) has been critically investigated for the past two decades, and novel findings regarding MC4R signalling and its potential exploitation in weight loss therapy have lately been emphasized. An association between MC4R and obesity is well established, with disease-causing mutations affecting 1% to 6% of obese patients. More than 200 MC4R variants have been reported, although conflicting results as to their effects have been found in different cohorts. Most notably, some MC4R gain-of-function variants seem to rescue obesity and related complications via specific pathways such as beta-arrestin (ß-arrestin) recruitment. Broadly speaking, however, dysfunctional MC4R dysregulates satiety and induces hyperphagia. The picture at the mechanistic level is complicated as, in addition to the canonical G stimulatory pathway, the ß-arrestin signalling pathway and ions (particularly calcium) seem to interact with MC4R signalling to contribute to or alleviate obesity pathogenesis. Thus, the overall complexity of the MC4R signalling spectra has broadened considerably, indicating there is great potential for the development of new drugs to manage obesity and its related complications. Alpha-melanocyte-stimulating hormone is the major endogenous MC4R agonist, but structure-based ligand discovery studies have identified possible superior and selective agonists that can improve MC4R function. However, some of these agonists characterized in vitro and in vivo confer adverse effects in patients, as demonstrated in clinical trials. In this review, we provide a comprehensive insight into the genetics, function and regulation of MC4R and its contribution to obesity. We also outline new approaches in drug development and emerging drug candidates to treat obesity.
Collapse
Affiliation(s)
- Munazza Tamkeen Fatima
- Department of Human Genetics, Translational Medicine DivisionResearch Branch, Sidra MedicineDohaQatar
| | - Ikhlak Ahmed
- Department of Human Genetics, Translational Medicine DivisionResearch Branch, Sidra MedicineDohaQatar
| | - Khalid Adnan Fakhro
- Department of Human Genetics, Translational Medicine DivisionResearch Branch, Sidra MedicineDohaQatar
- Department of Genetic MedicineWeill Cornell MedicineDohaQatar
- College of Health and Life SciencesHamad Bin Khalifa UniversityDohaQatar
| | | |
Collapse
|
10
|
Ahamed SK, Barek MA, Roy UK, Kouser M, Reza MS, Mannan AB, Alam MA, Uddin SMN. A review on association and correlation of genetic variants with eating disorders and obesity. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2021. [DOI: 10.1186/s43094-021-00250-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Now, eating disorders and obesity and their correlations are danger signal in worldwide which is caused by multifactor and associated with significant mortality and morbidity.
Main body
Every aspect of a patient’s life is influenced by eating disorders and obesity and their correlations. Due to frequent seeing of obese patients, eating disorders have been included in the review as they can sometimes be associated with obesity. However, it should be noted that most patients having eating disorder are at risk to be obese or overweight. This research explores the risk factors for the two disorders, as well as the assessment of medical complications and treatment recommendations for the disorders. In these two disorders, there is also a correlation. The essential consideration is that eating disorders are impulse-control disorders which are similar to addictive behaviors in some aspects. So it is a crying need to treat a patient with obesity and eating disorders simultaneously to ensure success. Genome-wide association studies (GWASs) have increased our knowledge of the pathophysiology of eating disorders (EDs) and obesity and their correlation.
Conclusion
This review enlightens on the summary of eating disorder, obesity, genotypic traits, molecular relations, interaction, correlation, and effect of eating disorder and obesity which outline potential future directions and clinical implications for patients with EDs and obesity.
Collapse
|
11
|
Adamska-Patruno E, Bauer W, Bielska D, Fiedorczuk J, Moroz M, Krasowska U, Czajkowski P, Wielogorska M, Maliszewska K, Puckowska S, Szczerbinski L, Lipinska D, Gorska M, Kretowski A. An Association between Diet and MC4R Genetic Polymorphism, in Relation to Obesity and Metabolic Parameters-A Cross Sectional Population-Based Study. Int J Mol Sci 2021; 22:ijms222112044. [PMID: 34769477 PMCID: PMC8584592 DOI: 10.3390/ijms222112044] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/27/2021] [Accepted: 11/05/2021] [Indexed: 11/30/2022] Open
Abstract
The melanocortin-4 receptor (MC4R) gene harbours one of the strongest susceptibility loci for obesity and obesity-related metabolic consequences. We analysed whether dietary factors may attenuate the associations between MC4R genotypes and obesity and metabolic parameters. In 819 participants genotyped for common MC4R polymorphisms (rs17782313, rs12970134, rs633265, and rs135034), the anthropometric measurements, body fat content and distribution (visceral and subcutaneous adipose tissue, VAT and SAT, respectively), and blood glucose, insulin, total-, LDL-, HDL-cholesterol, triglycerides concentrations, and daily macronutrient intake were assessed. ANOVA or Kruskal–Wallis tests were used, and multivariate linear regression models were developed. We observed that the CC genotype carriers (rs17782313) presented higher VAT, VAT/SAT ratio, fasting blood glucose and triglyceride concentrations when they were stratified to the upper quantiles of protein intake. An increase in energy derived from proteins was associated with higher BMI (Est. 5.74, R2 = 0.12), body fat content (Est. 8.44, R2 = 0.82), VAT (Est. 32.59, R2 = 0.06), and VAT/SAT ratio (Est. 0.96, R2 = 0.05). The AA genotype carriers (rs12970134) presented higher BMI, body fat, SAT and VAT, fasting blood glucose, triglycerides and total cholesterol concentrations. An increase in energy derived from proteins by AA carriers was associated with higher VAT (Est.19.95, R2 = 0.06) and VAT/SAT ratio (Est. 0.64, R2 = 0.05). Our findings suggest that associations of the common MC4R SNPs with obesity and its metabolic complications may be dependent on the daily dietary intake, which may open new areas for developing personalised diets for preventing and treating obesity and obesity-related comorbidities.
Collapse
Affiliation(s)
- Edyta Adamska-Patruno
- Clinical Research Centre, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (W.B.); (U.K.); (P.C.); (S.P.); (L.S.); (A.K.)
- Correspondence:
| | - Witold Bauer
- Clinical Research Centre, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (W.B.); (U.K.); (P.C.); (S.P.); (L.S.); (A.K.)
| | - Dorota Bielska
- Department of Family Medicine, Medical University of Bialystok, Mieszka I 4b, 15-054 Bialystok, Poland;
| | - Joanna Fiedorczuk
- Clinical Research Centre, Medical University of Bialystok Clinical Hospital, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (J.F.); (M.M.)
| | - Monika Moroz
- Clinical Research Centre, Medical University of Bialystok Clinical Hospital, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (J.F.); (M.M.)
| | - Urszula Krasowska
- Clinical Research Centre, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (W.B.); (U.K.); (P.C.); (S.P.); (L.S.); (A.K.)
| | - Przemyslaw Czajkowski
- Clinical Research Centre, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (W.B.); (U.K.); (P.C.); (S.P.); (L.S.); (A.K.)
| | - Marta Wielogorska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (M.W.); (K.M.); (D.L.); (M.G.)
| | - Katarzyna Maliszewska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (M.W.); (K.M.); (D.L.); (M.G.)
| | - Sylwia Puckowska
- Clinical Research Centre, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (W.B.); (U.K.); (P.C.); (S.P.); (L.S.); (A.K.)
| | - Lukasz Szczerbinski
- Clinical Research Centre, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (W.B.); (U.K.); (P.C.); (S.P.); (L.S.); (A.K.)
| | - Danuta Lipinska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (M.W.); (K.M.); (D.L.); (M.G.)
| | - Maria Gorska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (M.W.); (K.M.); (D.L.); (M.G.)
| | - Adam Kretowski
- Clinical Research Centre, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (W.B.); (U.K.); (P.C.); (S.P.); (L.S.); (A.K.)
- Clinical Research Centre, Medical University of Bialystok Clinical Hospital, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (J.F.); (M.M.)
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, Marii Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland; (M.W.); (K.M.); (D.L.); (M.G.)
| |
Collapse
|
12
|
Koerber-Rosso I, Brandt S, von Schnurbein J, Fischer-Posovszky P, Hoegel J, Rabenstein H, Siebert R, Wabitsch M. A fresh look to the phenotype in mono-allelic likely pathogenic variants of the leptin and the leptin receptor gene. Mol Cell Pediatr 2021; 8:10. [PMID: 34448070 PMCID: PMC8390564 DOI: 10.1186/s40348-021-00119-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Accepted: 07/19/2021] [Indexed: 12/16/2022] Open
Abstract
Leptin (LEP) and leptin receptor (LEPR) play a major role in energy homeostasis, metabolism, and reproductive function. While effects of biallelic likely pathogenic variants (-/-) on the phenotype are well characterized, effects of mono-allelic likely pathogenic variants (wt/-) in the LEP and LEPR gene on the phenotype compared to wild-type homozygosity (wt/wt) have not been systematically investigated. We identified in our systematic review 44 animal studies (15 on Lep, 29 on Lepr) and 39 studies in humans reporting on 130 mono-allelic likely pathogenic variant carriers with 20 distinct LEP variants and 108 heterozygous mono-allelic likely pathogenic variant carriers with 35 distinct LEPR variants. We found indications for a higher weight status in carriers of mono-allelic likely pathogenic variant in the leptin and in the leptin receptor gene compared to wt/wt, in both animal and human studies. In addition, animal studies showed higher body fat percentage in Lep and Lepr wt/- vs wt/wt. Animal studies provided indications for lower leptin levels in Lep wt/- vs. wt/wt and indications for higher leptin levels in Lepr wt/- vs wt/wt. Data on leptin levels in human studies was limited. Evidence for an impaired metabolism in mono-allelic likely pathogenic variants of the leptin and in leptin receptor gene was not conclusive (animal and human studies). Mono-allelic likely pathogenic variants in the leptin and in leptin receptor gene have phenotypic effects disposing to increased body weight and fat accumulation.
Collapse
Affiliation(s)
- Ingrid Koerber-Rosso
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Stephanie Brandt
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Julia von Schnurbein
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Pamela Fischer-Posovszky
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Josef Hoegel
- Institute of Human Genetics, University of Ulm, University Medical Center Ulm, Ulm, Germany
| | - Hannah Rabenstein
- Institute of Human Genetics, University of Ulm, University Medical Center Ulm, Ulm, Germany
| | - Reiner Siebert
- Institute of Human Genetics, University of Ulm, University Medical Center Ulm, Ulm, Germany
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany.
| |
Collapse
|
13
|
Zhang H, Chen LN, Yang D, Mao C, Shen Q, Feng W, Shen DD, Dai A, Xie S, Zhou Y, Qin J, Sun JP, Scharf DH, Hou T, Zhou T, Wang MW, Zhang Y. Structural insights into ligand recognition and activation of the melanocortin-4 receptor. Cell Res 2021; 31:1163-1175. [PMID: 34433901 DOI: 10.1038/s41422-021-00552-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022] Open
Abstract
Melanocortin-4 receptor (MC4R) plays a central role in the regulation of energy homeostasis. Its high sequence similarity to other MC receptor family members, low agonist selectivity and the lack of structural information concerning MC4R-specific activation have hampered the development of MC4R-seletive therapeutics to treat obesity. Here, we report four high-resolution structures of full-length MC4R in complex with the heterotrimeric Gs protein stimulated by the endogenous peptide ligand α-MSH, FDA-approved drugs afamelanotide (Scenesse™) and bremelanotide (Vyleesi™), and a selective small-molecule ligand THIQ, respectively. Together with pharmacological studies, our results reveal the conserved binding mode of peptidic agonists, the distinctive molecular details of small-molecule agonist recognition underlying receptor subtype selectivity, and a distinct activation mechanism for MC4R, thereby offering new insights into G protein coupling. Our work may facilitate the discovery of selective therapeutic agents targeting MC4R.
Collapse
Affiliation(s)
- Huibing Zhang
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China.,MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Li-Nan Chen
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China.,MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Dehua Yang
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Chunyou Mao
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China.,MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Qingya Shen
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China.,MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Wenbo Feng
- School of Pharmacy, Fudan University, Shanghai, China
| | - Dan-Dan Shen
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China.,MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Antao Dai
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Shanshan Xie
- Department of Cell Biology and Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Institute of Gastroenterology, Zhejiang University, Hangzhou, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang, China; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Yan Zhou
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China.,The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
| | - Jiao Qin
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China.,MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Laboratory of Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jin-Peng Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University; Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing, China.,Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Daniel H Scharf
- Department of Microbiology and The Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Tingjun Hou
- Innovation Institute for Artificial Intelligence in Medicine of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Tianhua Zhou
- Department of Cell Biology and Cancer Institute of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Institute of Gastroenterology, Zhejiang University, Hangzhou, China; Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Hangzhou, Zhejiang, China; Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Ming-Wei Wang
- The CAS Key Laboratory of Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China. .,The National Center for Drug Screening, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China. .,University of Chinese Academy of Sciences, Beijing, China. .,School of Pharmacy, Fudan University, Shanghai, China. .,School of Life Science and Technology, ShanghaiTech University, Shanghai, China. .,Department of Pharmacology, Fudan University, Shanghai, China.
| | - Yan Zhang
- Department of Biophysics and Department of Pathology of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China. .,Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, Zhejiang, China. .,MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China. .,Key Laboratory of Immunity and Inflammatory Diseases of Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
14
|
Harris SR, Carrillo M, Fujioka K. Binge-Eating Disorder and Type 2 Diabetes: A Review. Endocr Pract 2021; 27:158-164. [PMID: 33554873 DOI: 10.1016/j.eprac.2020.10.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 09/01/2020] [Accepted: 10/27/2020] [Indexed: 12/31/2022]
Abstract
OBJECTIVE To familiarize health care providers with diagnosis and treatment of binge-eating disorder (BED), a common comorbidity of type 2 diabetes (T2DM). METHODS Literature review of binge eating and T2DM. Key words used in search include BED, T2DM, obesity, and treatment. RESULTS The prevalence of BED in patients with T2DM appears to be much higher than the 2% to 3.5% prevalence seen in the general population. Studies suggest that up to 20% of patients with T2DM have an underlying eating disorder, the most common of which is binge eating. BED is probably underdiagnosed, even though there are multiple simple tools that providers can use to improve screening for the disorder. Though the relationship between BED and hemoglobin A1c control can vary, it appears that binge-eating behaviors can worsen metabolic markers, including glycemic control. Various medications used by patients with diabetes have been associated with new-onset BED, and treatment may be as simple as removing or replacing such agents. Several medications have been found to significantly reduce binge-eating frequency, and potentially, weight. Patients with BED generally benefit from psychotherapy, including cognitive behavioral therapy. CONCLUSION BED, only recently added to the International Classification of Disease-10 diagnostic list, is very common in patients with obesity and T2DM. The diagnosis is important to establish, as treatment or referral for treatment, could potentially improve many of the comorbidities and metrics of T2DM.
Collapse
Affiliation(s)
- Samantha R Harris
- Scripps Clinic Medical Group, Division of Diabetes and Endocrinology, La Jolla, California
| | - Maritza Carrillo
- Scripps Clinic Medical Group, Division of Diabetes and Endocrinology, La Jolla, California.
| | - Ken Fujioka
- Scripps Clinic Medical Group, Division of Diabetes and Endocrinology, La Jolla, California
| |
Collapse
|
15
|
Mason KE, Palla L, Pearce N, Phelan J, Cummins S. Genetic risk of obesity as a modifier of associations between neighbourhood environment and body mass index: an observational study of 335 046 UK Biobank participants. BMJ Nutr Prev Health 2021; 3:247-255. [PMID: 33521535 PMCID: PMC7841812 DOI: 10.1136/bmjnph-2020-000107] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/02/2020] [Accepted: 09/03/2020] [Indexed: 11/10/2022] Open
Abstract
Background There is growing recognition that recent global increases in obesity are the product of a complex interplay between genetic and environmental factors. However, in gene-environment studies of obesity, ‘environment’ usually refers to individual behavioural factors that influence energy balance, whereas more upstream environmental factors are overlooked. We examined gene-environment interactions between genetic risk of obesity and two neighbourhood characteristics likely to be associated with obesity (proximity to takeaway/fast-food outlets and availability of physical activity facilities). Methods We used data from 335 046 adults aged 40–70 in the UK Biobank cohort to conduct a population-based cross-sectional study of interactions between neighbourhood characteristics and genetic risk of obesity, in relation to body mass index (BMI). Proximity to a fast-food outlet was defined as distance from home address to nearest takeaway/fast-food outlet, and availability of physical activity facilities as the number of formal physical activity facilities within 1 km of home address. Genetic risk of obesity was operationalised by weighted Genetic Risk Scores of 91 or 69 single nucleotide polymorphisms (SNP), and by six individual SNPs considered separately. Multivariable, mixed-effects models with product terms for the gene-environment interactions were estimated. Results After accounting for likely confounding, the association between proximity to takeaway/fast-food outlets and BMI was stronger among those at increased genetic risk of obesity, with evidence of an interaction with polygenic risk scores (p=0.018 and p=0.028 for 69-SNP and 91-SNP scores, respectively) and in particular with a SNP linked to MC4R (p=0.009), a gene known to regulate food intake. We found very little evidence of gene-environment interaction for the availability of physical activity facilities. Conclusions Individuals at an increased genetic risk of obesity may be more sensitive to exposure to the local fast-food environment. Ensuring that neighbourhood residential environments are designed to promote a healthy weight may be particularly important for those with greater genetic susceptibility to obesity.
Collapse
Affiliation(s)
- Kate E Mason
- Department of Public Health, Policy and Systems, University of Liverpool, Liverpool, UK.,Department of Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Luigi Palla
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, UK.,Department of Global Health, University of Nagasaki, Nagasaki, Japan
| | - Neil Pearce
- Department of Medical Statistics, London School of Hygiene and Tropical Medicine, London, UK
| | - Jody Phelan
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Steven Cummins
- Department of Public Health, Environments and Society, London School of Hygiene and Tropical Medicine, London, UK
| |
Collapse
|
16
|
Wilson EA, Sun H, Cui Z, Jahnke MT, Pandey M, Metzger P, Gavrilova O, Chen M, Weinstein LS. G qα/G 11α deficiency in dorsomedial hypothalamus leads to obesity resulting from decreased energy expenditure and impaired sympathetic nerve activity. Am J Physiol Endocrinol Metab 2021; 320:E270-E280. [PMID: 33166186 PMCID: PMC8260363 DOI: 10.1152/ajpendo.00059.2020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The G-protein subunits Gqα and G11α (Gq/11α) couple receptors to phospholipase C, leading to increased intracellular calcium. In this study we investigated the consequences of Gq/11α deficiency in the dorsomedial hypothalamus (DMH), a critical site for the control of energy homeostasis. Mice with DMH-specific deletion of Gq/11α (DMHGq/11KO) were generated by stereotaxic injection of adeno-associated virus (AAV)-Cre-green fluorescent protein (GFP) into the DMH of Gqαflox/flox:G11α-/- mice. Compared with control mice that received DMH injection of AAV-GFP, DMHGq/11KO mice developed obesity associated with reduced energy expenditure without significant changes in food intake or physical activity. DMHGq/11KO mice showed no defects in the ability of the melanocortin agonist melanotan II to acutely stimulate energy expenditure or to inhibit food intake. At room temperature (22°C), DMHGq/11KO mice showed reduced sympathetic nervous system activity in brown adipose tissue (BAT) and heart, accompanied with decreased basal BAT uncoupling protein 1 (Ucp1) gene expression and lower heart rates. These mice were cold intolerant when acutely exposed to cold (6°C for 5 h) and had decreased cold-stimulated BAT Ucp1 gene expression. DMHGq/11KO mice also failed to adapt to gradually declining ambient temperatures and to develop adipocyte browning in inguinal white adipose tissue although their BAT Ucp1 was proportionally stimulated. Consistent with impaired cold-induced thermogenesis, the onset of obesity in DMHGq/11KO mice was significantly delayed when housed under thermoneutral conditions (30°C). Thus our results show that Gqα and G11α in the DMH are required for the control of energy homeostasis by stimulating energy expenditure and thermoregulation.NEW & NOTEWORTHY This paper demonstrates that signaling within the dorsomedial hypothalamus via the G proteins Gqα and G11α, which couple cell surface receptors to the stimulation of phospholipase C, is critical for regulation of energy expenditure, thermoregulation by brown adipose tissue and the induction of white adipose tissue browning.
Collapse
Affiliation(s)
- Eric A Wilson
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| | - Hui Sun
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| | - Zhenzhong Cui
- Mouse Metabolism Core Laboratory, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| | - Marshal T Jahnke
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| | - Mritunjay Pandey
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| | - Peter Metzger
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| | - Oksana Gavrilova
- Mouse Metabolism Core Laboratory, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| | - Min Chen
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| | - Lee S Weinstein
- Metabolic Diseases Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland
| |
Collapse
|
17
|
Santos JL, Cortés VA. Eating behaviour in contrasting adiposity phenotypes: Monogenic obesity and congenital generalized lipodystrophy. Obes Rev 2021; 22:e13114. [PMID: 33030294 DOI: 10.1111/obr.13114] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 07/13/2020] [Indexed: 12/11/2022]
Abstract
Most known types of nonsyndromic monogenic obesity are caused by rare mutations in genes of the leptin-melanocortin pathway controlling appetite and adiposity. In contrast, congenital generalized lipodystrophy represents the most extreme form of leanness in humans caused by recessive mutations in four genes involved in phospholipid/triglyceride synthesis and lipid droplet/caveolae structure. In this disease, the inability to store triglyceride in adipocytes results in hypoleptinemia and ectopic hepatic and muscle fat accumulation leading to fatty liver, hypertriglyceridemia and severe insulin resistance. As a result of hypoleptinemia, patients with lipodystrophy show alterations in eating behaviour characterized by constant increased energy intake. As it occurs in obesity caused by genetic leptin deficiency, exogenous leptin rapidly reduces hunger scores in patients with congenital generalized lipodystrophy, with additional beneficial effects on glucose homeostasis and metabolic profile normalization. The melanocortin-4 receptor agonist setmelanotide has been used in the treatment of monogenic obesities. There is only one report on the effect of setmelanotide in a patient with partial lipodystrophy resulting in mild reductions in hunger scores, with no improvements in metabolic status. The assessment of contrasting phenotypes of obesity/leanness represents an adequate strategy to understand the pathophysiology and altered eating behaviour associated with adipose tissue excessive accumulation/paucity.
Collapse
Affiliation(s)
- José L Santos
- Department of Nutrition, Diabetes and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Víctor A Cortés
- Department of Nutrition, Diabetes and Metabolism, School of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
18
|
Rare genetic forms of obesity: From gene to therapy. Physiol Behav 2020; 227:113134. [DOI: 10.1016/j.physbeh.2020.113134] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 07/15/2020] [Accepted: 07/21/2020] [Indexed: 01/05/2023]
|
19
|
Micioni Di Bonaventura E, Botticelli L, Tomassoni D, Tayebati SK, Micioni Di Bonaventura MV, Cifani C. The Melanocortin System behind the Dysfunctional Eating Behaviors. Nutrients 2020; 12:E3502. [PMID: 33202557 PMCID: PMC7696960 DOI: 10.3390/nu12113502] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 12/11/2022] Open
Abstract
The dysfunction of melanocortin signaling has been associated with obesity, given the important role in the regulation of energy homeostasis, food intake, satiety and body weight. In the hypothalamus, the melanocortin-3 receptor (MC3R) and melanocortin-4 receptor (MC4R) contribute to the stability of these processes, but MC3R and MC4R are also localized in the mesolimbic dopamine system, the region that responds to the reinforcing properties of highly palatable food (HPF) and where these two receptors seem to affect food reward and motivation. Loss of function of the MC4R, resulting from genetic mutations, leads to overeating in humans, but to date, a clear understanding of the underlying mechanisms and behaviors that promote overconsumption of caloric foods remains unknown. Moreover, the MC4R demonstrated to be a crucial modulator of the stress response, factor that is known to be strictly related to binge eating behavior. In this review, we will explore the preclinical and clinical studies, and the controversies regarding the involvement of melanocortin system in altered eating patterns, especially binge eating behavior, food reward and motivation.
Collapse
Affiliation(s)
| | - Luca Botticelli
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (E.M.D.B.); (L.B.); (S.K.T.); (C.C.)
| | - Daniele Tomassoni
- School of Bioscience and Veterinary Medicine, University of Camerino, 62032 Camerino, Italy;
| | - Seyed Khosrow Tayebati
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (E.M.D.B.); (L.B.); (S.K.T.); (C.C.)
| | | | - Carlo Cifani
- School of Pharmacy, University of Camerino, 62032 Camerino, Italy; (E.M.D.B.); (L.B.); (S.K.T.); (C.C.)
| |
Collapse
|
20
|
Hainer V, Aldhoon Hainerová I, Kunešová M, Taxová Braunerová R, Zamrazilová H, Bendlová B. Melanocortin pathways: suppressed and stimulated melanocortin-4 receptor (MC4R). Physiol Res 2020; 69:S245-S254. [PMID: 33094623 DOI: 10.33549/physiolres.934512] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Leptin-melanocortin pathway plays an essential role in the body weight regulation. Enhanced melanocortin signaling in the hypothalamus results in both decreased food intake and increased energy expenditure. The discovery of monogenic obesities with dysfunction of melanocortin-4 receptor (MC4R) greatly contributed to understanding of energy balance regulation. This review presents phenotypical characterization and prevalence of the MC4R gene mutations. Genome-wide association studies revealed that MC4R gene is significantly related not only to monogenic obesities but also to common obesity. An interaction of variants in the MC4R gene with fat mass and obesity associated (FTO) gene significantly increases the risk for obesity, particularly in adolescence. On the other hand, about 15 % of the MC4R gene variants result in a gain of function that protects against obesity and is associated with favorable metabolic profile. Long-term attempts to activate the MC4R have recently been finalized by a discovery of setmelanotide, a novel specific MC4R agonist that is devoid of untoward cardiovascular side-effects. The employment of specific MC4R agonists may open new horizons not only in the treatment of rare monogenic obesities but also in some common obesities where stimulation of MC4R could be achieved.
Collapse
Affiliation(s)
- V Hainer
- Obesity Management Center, Institute of Endocrinology, Prague, Czech Republic.
| | | | | | | | | | | |
Collapse
|
21
|
Baxter J, Armijo PR, Flores L, Krause C, Samreen S, Tanner T. Updates on Monogenic Obesity in a Multifactorial Disease. Obes Surg 2020; 29:4077-4083. [PMID: 31641982 DOI: 10.1007/s11695-019-04200-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Obesity is a worldwide epidemic with rates nearly doubling over the last 30 years. Despite increasing prevalence, the multifactorial pathogenesis of obesity continues to be widely misunderstood. Investigating genetic drivers in the development of obesity is an important area of focus, as genetics move to the forefront of medicine and personalized treatment evolves. Thus, this narrative review focused on four genes which have genome-wide association study-documented links to obesity and obesity syndromes. We explored their involvement in the predisposition, progression, and prognosis of obesity. Leptin, leptin receptor, pro-opiomelanocortin, and melanocortin 4 receptor are our four genes of interest, and herein we elaborated on the current literature, pathogenesis, and available treatments for patients with these specific genetic mutations.
Collapse
Affiliation(s)
- Jared Baxter
- College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | | | - Laura Flores
- College of Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Crystal Krause
- Center for Advanced Surgical Technology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Sarah Samreen
- Center for Advanced Surgical Technology, University of Nebraska Medical Center, Omaha, NE, USA
- General Surgery, Department of Surgery, University of Nebraska Medical Center, 986245 Nebraska Medical Center, Omaha, NE, 68198-6245, USA
| | - Tiffany Tanner
- Center for Advanced Surgical Technology, University of Nebraska Medical Center, Omaha, NE, USA.
- General Surgery, Department of Surgery, University of Nebraska Medical Center, 986245 Nebraska Medical Center, Omaha, NE, 68198-6245, USA.
| |
Collapse
|
22
|
A review of binge eating disorder and obesity. NEUROPSYCHIATRIE : KLINIK, DIAGNOSTIK, THERAPIE UND REHABILITATION : ORGAN DER GESELLSCHAFT ÖSTERREICHISCHER NERVENÄRZTE UND PSYCHIATER 2020; 35:57-67. [PMID: 32346850 DOI: 10.1007/s40211-020-00346-w] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 03/24/2020] [Indexed: 12/13/2022]
Abstract
Binge eating disorder (BED) is a mental illness characterised by recurrent binge eating episodes in the absence of appropriate compensatory behaviours. Consequently, BED is strongly associated with obesity. The current review aims to provide an update of the most relevant aspects of BED (e.g., clinical profile, aetiology and treatment approaches), in order not only to facilitate a better understanding of the disorder and its clinical consequences, but also to identify potential targets of prevention and intervention. Patients with BED often present high comorbidity with other medical conditions and psychiatric disorders. Numerous risk factors have been associated with the development and maintenance of the disorder. Moreover, although some treatments for BED have proven to be effective in addressing different key aspects of the disorder, the rates of patients that have ever received specific treatment for BED are very low. The factors involved and how to implement effective treatments will be discussed for the purpose of addressing the eating symptomatology and comorbid obesity.
Collapse
|
23
|
Graham CAM, Pedlar CR, Hearne G, Lorente-Cebrián S, González-Muniesa P, Mavrommatis Y. The Association of Parental Genetic, Lifestyle, and Social Determinants of Health with Offspring Overweight. Lifestyle Genom 2020; 13:99-106. [PMID: 32069471 DOI: 10.1159/000505749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 01/03/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION In the UK, the number of comorbidities seen in children has increased along with the worsening obesity rate. These comorbidities worsen into adulthood. Genome-wide association studies have highlighted single nucleotide polymorphisms associated with the weight status of adults and offspring individually. To date, in the UK, parental genetic, lifestyle, and social determinants of health have not been investigated alongside one another as influencers of offspring weight status. A comprehensive obesity prevention scheme would commence prior to conception and involve parental intervention including all known risk factors. This current study aims to identify the proportion of overweight that can be explained by known parental risk factors, including genetic, lifestyle, and social determinants of health with offspring weight status in the UK. METHODS A cross-sectional study was carried out on 123 parents. Parental and offspring anthropometric data and parental lifestyle and social determinants of health data were self-reported. Parental genetic data were collected by use of GeneFiX saliva collection vials and genotype were assessed for brain-derived neurotrophic factor (BDNF) gene rs6265, melanocortin 4 receptor (MC4R) gene rs17782313, transmembrane protein 18 (TMEM18) gene rs2867125, and serine/threonine-protein kinase (TNN13K) gene rs1514175. Associations were assessed between parental data and the weight status of offspring. RESULTS Maternal body mass index modestly predicted child weight status (p < 0.015; R2 = 0.15). More mothers of overweight children carried the MC4R rs17782313 risk allele (77.8%; p = 0.007) compared to mothers of normal-weight children. Additionally, fathers who were not Caucasian and parents who slept for <7 h/night had a larger percentage of overweight children when compared to their counterparts (p = 0.039; p = 0.014, respectively). CONCLUSION Associations exist between the weight status of offspring based solely on parental genetic, lifestyle, and social determinants of health data. Further research is required to appropriately address future interventions based on genetic and lifestyle risk groups on a pre-parent cohort.
Collapse
Affiliation(s)
- Catherine A M Graham
- Faculty of Health and Life Sciences, Department of Sport, Health and Social Work, Oxford Brookes University, Oxford, United Kingdom,
| | - Charles R Pedlar
- Faculty of Sport Health and Applied Science, Department of Nutrition, St. Mary's University, Twickenham, London, United Kingdom
| | - Gary Hearne
- Faculty of Science and Technology, Department of Design Engineering and Mathematics, Middlesex University, London, United Kingdom
| | - Silvia Lorente-Cebrián
- Centre for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain.,IdiSNA - Navarra Health Research Institute, Pamplona, Spain.,CIBERobn Physiopathology of Obesity and Nutrition, Centre of Biomedical Research Network, ISCIII, Madrid, Spain
| | - Pedro González-Muniesa
- Department of Nutrition, Food Science and Physiology, School of Pharmacy and Nutrition, Pamplona, University of Navarra, Pamplona, Spain.,Centre for Nutrition Research, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain.,IdiSNA - Navarra Health Research Institute, Pamplona, Spain.,CIBERobn Physiopathology of Obesity and Nutrition, Centre of Biomedical Research Network, ISCIII, Madrid, Spain
| | - Yiannis Mavrommatis
- Faculty of Sport Health and Applied Science, Department of Nutrition, St. Mary's University, Twickenham, London, United Kingdom
| |
Collapse
|
24
|
Liu Q, Guo XN, Liu CY, Xu WH. A proposed synergistic effect of CSF1R and NMUR2 variants contributes to binge eating in hereditary diffuse leukoencephalopathy with spheroids. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:7. [PMID: 32055598 DOI: 10.21037/atm.2019.11.30] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background The genetic mechanisms of binge eating (BE) as a disease identity remain obscure. BE is usually viewed as a part of the behavioral variant of frontotemporal dementia (bvFTD) features. We encountered a family with hereditary diffuse leukoencephalopathy with spheroids (HDLS) that manifested uniformly with binge-eating-onset dementia. The genetic factors associated with the rare phenotype were investigated. Methods The detailed phenotypes of the patients were described. We performed whole-exome sequencing (WES) of family members and repeat-primed PCR to analyze the patients' expansion size of C9orf72, a well-established gene causing FTD. The WES results of additional HDLS patients without BE manifestations were also investigated. Results All affected individuals had a BE-dementia-epilepsy pattern of disease progression. A recurrent disease-causing mutation in CSF1R established the diagnosis of HDLS in the family. No abnormalities in the expansion size of C9orf72 were detected. The concurrence of a recurrent CSF1R mutation and a rare variant in NMUR2, a gene functionally related to BE, was revealed in the affected family members. No potentially pathogenic variants in other known BE-associated genes were identified. Both the NMUR2 variant and the CSF1R mutation cosegregated with the BE-dementia-epilepsy phenotype in the family. In three additional HDLS patients without BE, no pathogenic variants in NMUR2 were detected. Conclusions We propose that synergistic genetic effects of NMUR2 and CSF1R variants may exist and contribute to the development of the BE phenotype in HDLS. NMUR2 is one of the potential susceptible genes in BE and may contribute in a background of a disrupted structural neuronetwork. Further studies in other BE-related disorders are required.
Collapse
Affiliation(s)
- Qing Liu
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMCH), Beijing 100730, China
| | - Xia-Nan Guo
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMCH), Beijing 100730, China.,State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, CAMS & PUMCH, Beijing 100005, China.,Department of Nephrology, the First Affiliated Hospital of Dalian Medical University, Dalian Medical University, Dalian 116011, China
| | - Cai-Yan Liu
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMCH), Beijing 100730, China
| | - Wei-Hai Xu
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS & PUMCH), Beijing 100730, China
| |
Collapse
|
25
|
Ehtesham S, Qasim A, Meyre D. Loss-of-function mutations in the melanocortin-3 receptor gene confer risk for human obesity: A systematic review and meta-analysis. Obes Rev 2019; 20:1085-1092. [PMID: 31090190 DOI: 10.1111/obr.12864] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 03/20/2019] [Accepted: 03/21/2019] [Indexed: 02/06/2023]
Abstract
The association between rare coding loss-of-function (LOF) mutations in the melanocortin receptor 3 (MC3R) gene and human obesity is controversial. To fill this gap of knowledge, we performed a systematic review and meta-analysis of genetic association studies in all ages and ethnicities. Two reviewers independently performed risk of bias assessment and extracted data. We searched Medline, Embase, Web of Science Core Collection, BIOSIS Preview, CINAHL, ProQuest Dissertations & Theses, and reference lists of relevant studies. All case-control, cross-sectional, prospective, and retrospective studies that evaluated prevalence of rare (less than 1% frequency) coding partial/complete LOF mutations in MC3R among individuals with obesity and normal weight were included. Our systematic search identified 1925 references relevant to the present review. Six studies were deemed eligible. Meta-analysis of 2969 individuals with obesity and 2572 with normal weight showed a positive association between rare heterozygous coding partial/complete LOF mutations in MC3R and obesity in children and adults of European, North African, and Asian ancestries (odds ratio = 3.07; 95% CI, 1.48-7.00; P = 4.2 × 10-3 ). Our data demonstrates that rare partial/complete LOF mutations in the coding region of MC3R confer three-time increased risk of obesity in humans, and implies that rare genetic variants with intermediate effects contribute to the missing heritability of obesity.
Collapse
Affiliation(s)
- Sahar Ehtesham
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Anila Qasim
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
| | - David Meyre
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
26
|
Adamska-Patruno E, Goscik J, Czajkowski P, Maliszewska K, Ciborowski M, Golonko A, Wawrusiewicz-Kurylonek N, Citko A, Waszczeniuk M, Kretowski A, Gorska M. The MC4R genetic variants are associated with lower visceral fat accumulation and higher postprandial relative increase in carbohydrate utilization in humans. Eur J Nutr 2019; 58:2929-2941. [PMID: 30945034 PMCID: PMC6768895 DOI: 10.1007/s00394-019-01955-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 03/24/2019] [Indexed: 12/14/2022]
Abstract
Purpose The interactions between lifestyle and genetic factors play an important role in obesity development. Mutations in melanocortin-4-receptor (MC4R) gene are one of the most common cause of monogenic obesity, however, the functional effects of polymorphic variants near MC4R gene in general populations remain uncertain. The aim of our study was to analyze whether the common single nucleotide polymorphisms (SNPs) of MC4R gene influence the food preferences, physical activity, body fat content and distribution, as well as fasting and postprandial energy expenditure and substrates utilization. Methods We genotyped previously identified MC4R SNPs: rs17782313, rs633265, rs1350341, rs12970134 in 927 subjects, who underwent anthropometric, total body fat content, visceral (VAT) and subcutaneous adipose tissue (SAT) measurements, and daily physical activity and dietary intake analysis. In randomly selected 47 subjects the energy expenditure, carbohydrate and lipid utilizations were evaluated in fasting state and after high-carbohydrate and control meals intake. Results We found the significant associations between studied SNPs of MC4R gene and VAT and VAT/SAT ratio. Moreover, the GG genotype carriers of rs1350341, who had the lowest VAT accumulation (p = 0.012), presented higher relative increase in postprandial carbohydrate utilization (p = 0.013, p = 0.024). Conclusions We have observed that common SNPs of the MC4R gene influence the body fat content and distribution, as well as relative increase in postprandial carbohydrate utilization. We believe that our study may help to understand better the impact of MC4R gene on obesity development, and to help to provide personalized prevention/treatment strategies to fight against obesity and its metabolic consequences.
Collapse
Affiliation(s)
- Edyta Adamska-Patruno
- Clinical Research Centre, Medical University of Bialystok, M.C. Sklodowskiej-Curie 24A, 15-276, Bialystok, Poland.
| | - Joanna Goscik
- Centre for Experimental Medicine, Medical University of Bialystok, M.C. Sklodowskiej-Curie 24A, 15-276, Bialystok, Poland
| | - Przemyslaw Czajkowski
- Clinical Research Centre, Medical University of Bialystok, M.C. Sklodowskiej-Curie 24A, 15-276, Bialystok, Poland
| | - Katarzyna Maliszewska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, M.C. Sklodowskiej-Curie 24A, 15-276, Bialystok, Poland
| | - Michał Ciborowski
- Clinical Research Centre, Medical University of Bialystok, M.C. Sklodowskiej-Curie 24A, 15-276, Bialystok, Poland
| | - Anna Golonko
- Department of Dietetics and Clinical Nutrition, Medical University of Bialystok, Mieszka I-go 4B, 15-054, Bialystok, Poland
| | - Natalia Wawrusiewicz-Kurylonek
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, M.C. Sklodowskiej-Curie 24A, 15-276, Bialystok, Poland
| | - Anna Citko
- Clinical Research Centre, Medical University of Bialystok, M.C. Sklodowskiej-Curie 24A, 15-276, Bialystok, Poland
| | - Magdalena Waszczeniuk
- Clinical Research Centre, Medical University of Bialystok, M.C. Sklodowskiej-Curie 24A, 15-276, Bialystok, Poland.,Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, M.C. Sklodowskiej-Curie 24A, 15-276, Bialystok, Poland
| | - Adam Kretowski
- Clinical Research Centre, Medical University of Bialystok, M.C. Sklodowskiej-Curie 24A, 15-276, Bialystok, Poland.,Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, M.C. Sklodowskiej-Curie 24A, 15-276, Bialystok, Poland
| | - Maria Gorska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, M.C. Sklodowskiej-Curie 24A, 15-276, Bialystok, Poland
| |
Collapse
|
27
|
Anderson EJP, Ghamari-Langroudi M, Cakir I, Litt MJ, Chen V, Reggiardo RE, Millhauser GL, Cone RD. Late onset obesity in mice with targeted deletion of potassium inward rectifier Kir7.1 from cells expressing the melanocortin-4 receptor. J Neuroendocrinol 2019; 31:e12670. [PMID: 30561082 PMCID: PMC6533113 DOI: 10.1111/jne.12670] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 11/21/2018] [Accepted: 12/11/2018] [Indexed: 01/01/2023]
Abstract
Energy stores in fat tissue are determined in part by the activity of hypothalamic neurones expressing the melanocortin-4 receptor (MC4R). Even a partial reduction in MC4R expression levels in mice, rats or humans produces hyperphagia and morbid obesity. Thus, it is of great interest to understand the molecular basis of neuromodulation by the MC4R. The MC4R is a G protein-coupled receptor that signals efficiently through GαS , and this signalling pathway is essential for normal MC4R function in vivo. However, previous data from hypothalamic slice preparations indicated that activation of the MC4R depolarised neurones via G protein-independent regulation of the ion channel Kir7.1. In the present study, we show that deletion of Kcnj13 (ie, the gene encoding Kir7.1) specifically from MC4R neurones produced resistance to melanocortin peptide-induced depolarisation of MC4R paraventricular nucleus neurones in brain slices, resistance to the sustained anorexic effect of exogenously administered melanocortin peptides, late onset obesity, increased linear growth and glucose intolerance. Some MC4R-mediated phenotypes appeared intact, including Agouti-related peptide-induced stimulation of food intake and MC4R-mediated induction of peptide YY release from intestinal L cells. Thus, a subset of the consequences of MC4R signalling in vivo appears to be dependent on expression of the Kir7.1 channel in MC4R cells.
Collapse
Affiliation(s)
- E. J. P. Anderson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee
| | - M. Ghamari-Langroudi
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee
| | - I. Cakir
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan
| | - M. J. Litt
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee
| | - Valerie Chen
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, California
| | - Roman E. Reggiardo
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, California
| | - Glenn L. Millhauser
- Department of Chemistry and Biochemistry, University of California Santa Cruz, Santa Cruz, California
| | - R. D. Cone
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan
- Department of Molecular and Integrative Pharmacology, School of Medicine, University of Michigan, Ann Arbor, Michigan
- Correspondence: Roger D. Cone, Life Sciences Institute, 210 Washtenaw Ave., Ann Arbor, MI 48109,
| |
Collapse
|
28
|
da Fonseca ACP, Abreu GDM, Zembrzuski VM, Junior MC, Carneiro JRI, Magno FCCM, Rosado EL, Nogueira Neto JF, de Cabello GMK, Cabello PH. Identification of the MC4R start lost mutation in a morbidly obese Brazilian patient. Diabetes Metab Syndr Obes 2019; 12:257-266. [PMID: 30863132 PMCID: PMC6388727 DOI: 10.2147/dmso.s189455] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Melanocortin 4 receptor gene (MC4R) is an important regulator of food intake, body weight, and blood pressure. Mutations in MC4R are associated with the most common form of nonsyndromic monogenic obesity. MC4R variations have an autosomal co-/dominant model of inheritance. MC4R screening could reveal individuals previously unrecognized with Mendelian form of obesity for further clinical management and genetic counseling. However, there are limited data regarding MC4R variants in patients with obesity from Brazil. The aim of this study was to screen the coding region of the MC4R gene in a Brazilian cohort of severely obese adults and to investigate the phenotype-genotype correlation within MC4R variant carriers. METHODS This study comprised 157 adult participants, stratified according to the period of obesity onset. The first group included 97 patients with childhood-onset obesity (0-11 years) and the second group comprised 60 subjects with adolescence/youth-onset obesity (12-21 years). The entire coding region of MC4R gene was screened by Sanger sequencing. RESULTS As a result, five previously described variants (Met1?, Ser36Thr, Val103Ile, Ile98=, and Phe202Leu) were identified. Met1? is a start lost codon variant, which affects the translation of MC4R. It was found in a female patient with childhood-onset obesity. We also compared the anthropometric and metabolic parameters between patients with MC4R missense variants (Ser36Thr, Val103Ile, and Phe202Leu) and noncarriers. Patients carrying MC4R variants had higher median of waist-hip ratio when compared to noncarriers (P=0.048). These missense variants were also associated with hypertension (P=0.014). Additionally, Val103Ile carriers had lower diastolic blood pressure and lower systolic blood pressure compared to noncarriers (P=0.020 and P=0.065, respectively). Val103Ile was also associated with hypertension (P=0.003). CONCLUSION This study showed the prevalence of MC4R variants in a cohort of Brazilian adults with severe obesity. We also identified significant phenotype differences between carriers and noncarriers of missense variants in our sample, suggesting an important role of MC4R on body fat distribution and blood pressure.
Collapse
Affiliation(s)
| | - Gabriella de Medeiros Abreu
- Human Genetics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil,
| | - Verônica Marques Zembrzuski
- Human Genetics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil,
| | - Mario Campos Junior
- Human Genetics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil,
| | - João Regis Ivar Carneiro
- Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Eliane Lopes Rosado
- Institute of Nutrition Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | | | - Pedro Hernán Cabello
- Human Genetics Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (FIOCRUZ), Rio de Janeiro, Brazil,
- Human Genetics Laboratory, Grande Rio University, Rio de Janeiro, Brazil
| |
Collapse
|
29
|
Qasim A, Mayhew AJ, Ehtesham S, Alyass A, Volckmar AL, Herpertz S, Hinney A, Hebebrand J, Meyre D. Gain-of-function variants in the melanocortin 4 receptor gene confer susceptibility to binge eating disorder in subjects with obesity: a systematic review and meta-analysis. Obes Rev 2019; 20:13-21. [PMID: 30306707 DOI: 10.1111/obr.12761] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 07/20/2018] [Accepted: 08/02/2018] [Indexed: 01/21/2023]
Abstract
The association between coding variants in the melanocortin 4 receptor gene (MC4R) and binge eating disorder (BED) in patients with obesity is controversial. Two independent reviewers systematically searched MEDLINE, Embase, PsycINFO, BIOSIS Previews, Web of Science Core Collection and Google Scholar up to February 2018, using terms describing the MC4R gene and BED. Six of 103 identified references were included. Studies examined associations between at least one coding variant/mutation in MC4R and BED and screened for BED as per the Diagnostic and Statistical Manual of Mental Disorders. Risk of bias was assessed using a modified version of the Q-Genie tool, and overall quality of evidence was assessed using Grading of Recommendations Assessment, Development and Evaluation guidance. Meta-analysis was conducted via logistic regression models. A positive association between gain-of-function (GOF) variants in the MC4R and BED was observed (odds ratio [OR] = 3.05; 95% confidence interval [CI]: 1.82, 5.04; p = 1.7 × 10-5 ), while no association was detected between loss-of-function (LOF) mutations and BED (OR = 1.50; 95% CI: 0.73, 2.96; p = 0.25). Similar results were found after accounting for study quality (GOF variants: OR = 3.15; 95% CI: 1.76, 5.66; p = 1.1 × 10-4 ; LOF mutations: OR = 1.50; 95% CI: 0.73, 2.97; p = 0.25). Our systematic review and meta-analysis provides evidence that GOF variants as opposed to LOF mutations in MC4R are associated with BED in subjects with obesity.
Collapse
Affiliation(s)
- A Qasim
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - A J Mayhew
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - S Ehtesham
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - A Alyass
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada
| | - A-L Volckmar
- Department of Child and Adolescent Psychiatry, University Hospital Essen, University of Duisburg - Essen, Essen, Germany
| | - S Herpertz
- Department of Psychosomatic Medicine and Psychotherapy, LWL University Clinic, Ruhr University Bochum, Bochum, Germany
| | - A Hinney
- Department of Child and Adolescent Psychiatry, University Hospital Essen, University of Duisburg - Essen, Essen, Germany
| | - J Hebebrand
- Department of Child and Adolescent Psychiatry, University Hospital Essen, University of Duisburg - Essen, Essen, Germany
| | - D Meyre
- Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, ON, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW This review aims to present current information on genes underlying severe obesity, with the main emphasis on the three genes LEP, LEPR and MC4R. RECENT FINDINGS There is a substantial amount of evidence that variants in at least ten different genes are the cause of severe monogenic obesity. The majority of these are involved in the leptin-melanocortin signalling pathway. Due to the frequency of some of the identified variants, it is clear that monogenic variants also make a significant contribution to common obesity. The artificial distinction between rare monogenic obesity and common polygenic obesity is now obsolete with the identification of MC4R variants of strong effect in the general population.
Collapse
Affiliation(s)
- Una Fairbrother
- School of Human Sciences, London Metropolitan University, North Campus, 166-220 Holloway Road, London, N7 8DB, UK
| | - Elliot Kidd
- School of Human Sciences, London Metropolitan University, North Campus, 166-220 Holloway Road, London, N7 8DB, UK
| | - Tanya Malagamuwa
- Institute of Medical and Biomedical Education, St George's University of London, Cranmer Terrace, Tooting, London, SW17 0RE, UK
| | - Andrew Walley
- Institute of Medical and Biomedical Education, St George's University of London, Cranmer Terrace, Tooting, London, SW17 0RE, UK.
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW Genetic obesity is responsible for up to 7% of severe childhood obesity. Although current Pediatric Endocrine Society guidelines recommend assessment of children with early-onset morbid obesity and hyperphagia for underlying genetic disorders, a vast majority of patients are not being appropriately screened for genetic obesity syndromes. RECENT FINDINGS With advances in genetic testing, more genetic causes of obesity are being identified. Treatments are likely to be individualized, depending on the cause of the obesity, and must be targeted at addressing the underlying cause. Investigational therapies include melanocortin-4 receptor antagonists, oxytocin and medications targeting the endocannabinoid system. SUMMARY Improved identification of patients with genetic obesity syndromes will lead to development of new treatments and personalized management of these diseases.
Collapse
|
32
|
Iepsen EW, Zhang J, Thomsen HS, Hansen EL, Hollensted M, Madsbad S, Hansen T, Holst JJ, Holm JC, Torekov SS. Patients with Obesity Caused by Melanocortin-4 Receptor Mutations Can Be Treated with a Glucagon-like Peptide-1 Receptor Agonist. Cell Metab 2018; 28:23-32.e3. [PMID: 29861388 DOI: 10.1016/j.cmet.2018.05.008] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 03/09/2018] [Accepted: 05/05/2018] [Indexed: 01/12/2023]
Abstract
Pathogenic mutations in the appetite-regulating melanocortin-4 receptor (MC4R) represent the most common cause of monogenic obesity with limited treatment options. Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) cause weight loss by reducing appetite. We assessed the effect of the GLP-1 RA liraglutide 3.0 mg for 16 weeks in 14 obese individuals with pathogenic MC4R mutations (BMI 37.5 ± 6.8) and 28 matched control participants without MC4R mutation (BMI 36.8 ± 4.8). Liraglutide decreased body weight by 6.8 kg ± 1.8 kg in individuals with pathogenic MC4R mutations and by 6.1 kg ± 1.2 kg in control participants. Total body fat, waist circumference, and fasting and postprandial glucose concentrations similarly decreased in both groups. Thus, liraglutide induced an equal, clinically significant weight loss of 6% in both groups, indicating that the appetite-reducing effect of liraglutide is preserved in MC4R causal obesity and that liraglutide acts independently of the MC4R pathway. Thus, liraglutide could be an effective treatment of the most common form of monogenic obesity.
Collapse
Affiliation(s)
- Eva W Iepsen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; The Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Translational Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark.
| | - Jinyi Zhang
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; The Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Translational Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Henrik S Thomsen
- Department of Radiology, Herlev University Hospital, Herlev 2730, Denmark
| | - Elizaveta L Hansen
- Department of Radiology, Herlev University Hospital, Herlev 2730, Denmark
| | - Mette Hollensted
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Sten Madsbad
- Department of Endocrinology, Hvidovre University Hospital, Hvidovre 2650, Denmark
| | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Jens J Holst
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; The Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Translational Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Jens-Christian Holm
- The Children's Obesity Clinic, Department of Pediatrics, Holbæk University Hospital, Holbæk 4300, Denmark
| | - Signe S Torekov
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; The Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Translational Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark.
| |
Collapse
|
33
|
Bonnefond A, Keller R, Meyre D, Stutzmann F, Thuillier D, Stefanov DG, Froguel P, Horber FF, Kral JG. Eating Behavior, Low-Frequency Functional Mutations in the Melanocortin-4 Receptor (MC4R) Gene, and Outcomes of Bariatric Operations: A 6-Year Prospective Study. Diabetes Care 2016; 39:1384-92. [PMID: 27222505 DOI: 10.2337/dc16-0115] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 05/02/2016] [Indexed: 02/03/2023]
Abstract
OBJECTIVE Data on the effects of eating behavior and genetics on outcomes of gastrointestinal surgery for diabesity have been sparse, often flawed, and controversial. We aimed to assess long-term outcomes of bariatric operations in patients characterized for eating behavior and rare mutations in the melanocortin-4 receptor (MC4R) gene, which is strongly implicated in energy balance. RESEARCH DESIGN AND METHODS Between 1996 and 2005, 1,264 severely obese Swiss patients underwent current laparoscopic adjustable gastric banding, gastroduodenal bypass, or a hybrid operation. Of these, 872 patients were followed for a minimum of 6 years and were screened for MC4R mutations. Using regression models, we studied relationships between eating behavior and MC4R mutations and postoperative weight loss, complications, and reoperations after 6 years. RESULTS At baseline, rare functional MC4R mutation carriers exhibited a significantly higher prevalence of binge eating disorder (BED) or loss-of-control eating independent of age, sex, and BMI. Six years after bariatric surgery, the mutation carriers had more major complications than wild-type subjects independent of age, baseline BMI, sex, operation type, and weight loss. Furthermore, high baseline BMI, male sex, BED, and functional MC4R mutations were independent predictors of higher reoperation rates. CONCLUSIONS Sequencing of MC4R and eating typology, combined with stratification for sex and baseline BMI, might significantly improve patient allocation to banding or bypass operations for diabesity as well as reduce both complication and reoperation rates.
Collapse
Affiliation(s)
- Amélie Bonnefond
- CNRS UMR 8199, Lille Pasteur Institute, Lille, France Lille University, Lille, France European Genome Institute for Diabetes, FR 3508, Lille, France Department of Genomics of Common Disease, School of Public Health, Hammersmith Hospital, Imperial College London, London, U.K
| | - Ramsi Keller
- Department of Internal Medicine, Landesspital Liechtenstein, Vaduz, Liechtenstein Dr. Horber Adipositas Stiftung, Zurich, Switzerland
| | - David Meyre
- Department of Clinical Epidemiology and Biostatistics, McMaster University, Hamilton, Ontario, Canada Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Fanny Stutzmann
- CNRS UMR 8199, Lille Pasteur Institute, Lille, France Lille University, Lille, France European Genome Institute for Diabetes, FR 3508, Lille, France
| | - Dorothée Thuillier
- CNRS UMR 8199, Lille Pasteur Institute, Lille, France Lille University, Lille, France European Genome Institute for Diabetes, FR 3508, Lille, France
| | - Dimitre G Stefanov
- Scientific Computing Center and Departments of Surgery, Medicine, and Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY
| | - Philippe Froguel
- CNRS UMR 8199, Lille Pasteur Institute, Lille, France Lille University, Lille, France European Genome Institute for Diabetes, FR 3508, Lille, France Department of Genomics of Common Disease, School of Public Health, Hammersmith Hospital, Imperial College London, London, U.K.
| | - Fritz F Horber
- Department of Internal Medicine, Landesspital Liechtenstein, Vaduz, Liechtenstein Dr. Horber Adipositas Stiftung, Zurich, Switzerland University of Bern, Bern, Switzerland
| | - John G Kral
- Scientific Computing Center and Departments of Surgery, Medicine, and Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY
| |
Collapse
|
34
|
Lemche E, Chaban OS, Lemche AV. Neuroendocrinological and Epigenetic Mechanisms Subserving Autonomic Imbalance and HPA Dysfunction in the Metabolic Syndrome. Front Neurosci 2016; 10:142. [PMID: 27147943 PMCID: PMC4830841 DOI: 10.3389/fnins.2016.00142] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 03/21/2016] [Indexed: 12/18/2022] Open
Abstract
Impact of environmental stress upon pathophysiology of the metabolic syndrome (MetS) has been substantiated by epidemiological, psychophysiological, and endocrinological studies. This review discusses recent advances in the understanding of causative roles of nutritional factors, sympathomedullo-adrenal (SMA) and hypothalamic-pituitary adrenocortical (HPA) axes, and adipose tissue chronic low-grade inflammation processes in MetS. Disturbances in the neuroendocrine systems for leptin, melanocortin, and neuropeptide Y (NPY)/agouti-related protein systems have been found resulting directly in MetS-like conditions. The review identifies candidate risk genes from factors shown critical for the functioning of each of these neuroendocrine signaling cascades. In its meta-analytic part, recent studies in epigenetic modification (histone methylation, acetylation, phosphorylation, ubiquitination) and posttranscriptional gene regulation by microRNAs are evaluated. Several studies suggest modification mechanisms of early life stress (ELS) and diet-induced obesity (DIO) programming in the hypothalamic regions with populations of POMC-expressing neurons. Epigenetic modifications were found in cortisol (here HSD11B1 expression), melanocortin, leptin, NPY, and adiponectin genes. With respect to adiposity genes, epigenetic modifications were documented for fat mass gene cluster APOA1/C3/A4/A5, and the lipolysis gene LIPE. With regard to inflammatory, immune and subcellular metabolism, PPARG, NKBF1, TNFA, TCF7C2, and those genes expressing cytochrome P450 family enzymes involved in steroidogenesis and in hepatic lipoproteins were documented for epigenetic modifications.
Collapse
Affiliation(s)
- Erwin Lemche
- Section of Cognitive Neuropsychiatry, Department of Psychosis Studies, Institute of Psychiatry, Psychology and Neuroscience, King's College London London, UK
| | - Oleg S Chaban
- Section of Psychosomatic Medicine, Bogomolets National Medical University Kiev, Ukraine
| | - Alexandra V Lemche
- Department of Medical Science, Institute of Clinical Research Berlin, Germany
| |
Collapse
|
35
|
Donkin I, Versteyhe S, Ingerslev LR, Qian K, Mechta M, Nordkap L, Mortensen B, Appel EVR, Jørgensen N, Kristiansen VB, Hansen T, Workman CT, Zierath JR, Barrès R. Obesity and Bariatric Surgery Drive Epigenetic Variation of Spermatozoa in Humans. Cell Metab 2016; 23:369-78. [PMID: 26669700 DOI: 10.1016/j.cmet.2015.11.004] [Citation(s) in RCA: 329] [Impact Index Per Article: 41.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 08/27/2015] [Accepted: 11/04/2015] [Indexed: 02/06/2023]
Abstract
Obesity is a heritable disorder, with children of obese fathers at higher risk of developing obesity. Environmental factors epigenetically influence somatic tissues, but the contribution of these factors to the establishment of epigenetic patterns in human gametes is unknown. Here, we hypothesized that weight loss remodels the epigenetic signature of spermatozoa in human obesity. Comprehensive profiling of the epigenome of sperm from lean and obese men showed similar histone positioning, but small non-coding RNA expression and DNA methylation patterns were markedly different. In a separate cohort of morbidly obese men, surgery-induced weight loss was associated with a dramatic remodeling of sperm DNA methylation, notably at genetic locations implicated in the central control of appetite. Our data provide evidence that the epigenome of human spermatozoa dynamically changes under environmental pressure and offers insight into how obesity may propagate metabolic dysfunction to the next generation.
Collapse
Affiliation(s)
- Ida Donkin
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Soetkin Versteyhe
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Lars R Ingerslev
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Kui Qian
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Mie Mechta
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Loa Nordkap
- University Department of Growth and Reproduction, Copenhagen University Hospital, Copenhagen 2100, Denmark
| | - Brynjulf Mortensen
- Steno Diabetes Center, Gentofte 2810, Denmark; Center for Diabetes Research, University of Copenhagen, Gentofte Hospital, Hellerup 2900, Denmark
| | - Emil Vincent R Appel
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Niels Jørgensen
- University Department of Growth and Reproduction, Copenhagen University Hospital, Copenhagen 2100, Denmark
| | - Viggo B Kristiansen
- Department of Surgical Gastroenterology, Hvidovre Hospital, Hvidovre 2650, Denmark
| | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Christopher T Workman
- Department of Systems Biology, Technical University of Denmark, Lyngby 2800, Denmark; Center for non-coding RNA in Technology and Health, Faculty of Health and Medical Sciences, University of Copenhagen, Frederiksberg 1870, Denmark
| | - Juleen R Zierath
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark; Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm 171 77, Sweden
| | - Romain Barrès
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark.
| |
Collapse
|
36
|
Huvenne H, Dubern B, Clément K, Poitou C. Rare Genetic Forms of Obesity: Clinical Approach and Current Treatments in 2016. Obes Facts 2016; 9:158-73. [PMID: 27241181 PMCID: PMC5644891 DOI: 10.1159/000445061] [Citation(s) in RCA: 138] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 02/03/2016] [Indexed: 12/28/2022] Open
Abstract
Obesity results from a synergistic relationship between genes and the environment. The phenotypic expression of genetic factors involved in obesity is variable, allowing to distinguish several clinical pictures of obesity. Monogenic obesity is described as rare and severe early-onset obesity with abnormal feeding behavior and endocrine disorders. This is mainly due to autosomal recessive mutations in genes of the leptin-melanocortin pathway which plays a key role in the hypothalamic control of food intake. Melanocortin 4 receptor(MC4R)-linked obesity is characterized by the variable severity of obesity and no notable additional phenotypes. Mutations in the MC4R gene are involved in 2-3% of obese children and adults; the majority of these are heterozygous. Syndromic obesity is associated with mental retardation, dysmorphic features, and organ-specific developmental abnormalities. Additional genes participating in the development of hypothalamus and central nervous system have been regularly identified. But to date, not all involved genes have been identified so far. New diagnostic tools, such as whole-exome sequencing, will probably help to identify other genes. Managing these patients is challenging. Indeed, specific treatments are available only for specific types of monogenic obesity, such as leptin deficiency. Data on bariatric surgery are limited and controversial. New molecules acting on the leptin-melanocortin pathway are currently being developed.
Collapse
Affiliation(s)
- Hélène Huvenne
- GHICL, Saint-Vincent de Paul Hospital, Department of Pediatrics, Lille, France
| | | | | | | |
Collapse
|
37
|
Thakare MM, Surana SJ. β-Asarone modulate adipokines and attenuates high fat diet-induced metabolic abnormalities in Wistar rats. Pharmacol Res 2015; 103:227-35. [PMID: 26675715 DOI: 10.1016/j.phrs.2015.12.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 12/01/2015] [Accepted: 12/01/2015] [Indexed: 12/15/2022]
Abstract
Here we investigated the effect of β-asarone on food preference and its therapeutic potential against high fat diet (HFD) induced obesity in rats. In food preference study, free access to HFD was given only for 4h in addition to standard laboratory chow in rats and the preferential intake between chow and HFD was measured. For obesity induction, HFD was administered for 12 weeks and the HFD fed rats were treated with β-asarone in the last 4 weeks, starting from 9th week onwards. Food intake, body weight was measured biweekly. Glucose tolerance and the levels of glucose, lipids, free fatty acids, leptin, and adiponectin were assessed. HFD fed rats showed progressive increase in body weight and developed glucose intolerance and dyslipidemia. In addition, they showed increased adiposity and the disturbed pattern of adipokine levels In the food preference paradigm, β-asarone produced selective decrease in HFD intake in rats. In obese rats, β-asarone treatment not only reduced body weight but also prevented HFD-induced metabolic alterations, including glucose intolerance, dyslipidemia and adipokine imbalance. The observed beneficial effects of β-asarone appear due its ability to reduce intake of energy dense food by affecting food palatability, and to normalize the levels of leptin and adiponectin in rats. Overall, our results suggest that β-asarone is a novel candidate molecule with significant therapeutic potential in the management of obesity and associated abnormalities.
Collapse
Affiliation(s)
- Malesh M Thakare
- Department of Pharmacology, RC Patel Institute of Pharmaceutical Education and Research, Shirpur 425 405, Dhule, Maharashtra, India.
| | - Sanjay J Surana
- Department of Pharmacology, RC Patel Institute of Pharmaceutical Education and Research, Shirpur 425 405, Dhule, Maharashtra, India
| |
Collapse
|
38
|
Melanocortin-4 receptor gene variants are not associated with binge-eating behavior in nonobese patients with eating disorders. Psychiatr Genet 2015; 25:35-8. [PMID: 25419636 DOI: 10.1097/ypg.0000000000000065] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
We aimed to determine whether variability in the melanocortin-4 receptor (MC4R) gene, predisposing to hyperphagia and obesity, may also be present in nonobese patients with binge-eating behavior or be related to anthropometric or psychopathological parameters in these patients. The coding region of the MC4R gene was sequenced in nonobese patients with binge-eating behavior diagnosed with bulimia nervosa or binge-eating disorder (n=77); individuals with severe early-onset obesity (n=170); and lean women with anorexia nervosa (n=20). A psychometric evaluation (Eating Disorders Inventory-2 and Symptom Checklist 90 Revised inventories) was carried out for all the patients with eating disorders. In the obesity group, 10 different variants were identified, whereas in the binge-eating patients, only two individuals with bulimia nervosa were found to carry the I251L polymorphism, which did not correlate with weight, BMI, or psychopathological features. We found no evidence that mutations in the MC4R gene are associated with binge-eating behavior in nonobese eating disorder patients.
Collapse
|
39
|
Dar R, Rasool S, Zargar AH, Jan TR, Andrabi KI. Polymorphic analysis of MC4R gene in ethnic Kashmiri population with type 2 diabetes. Int J Diabetes Dev Ctries 2015. [DOI: 10.1007/s13410-015-0454-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
|
40
|
Young KL, Graff M, North KE, Richardson AS, Mohlke KL, Lange LA, Lange EM, Harris KM, Gordon-Larsen P. Interaction of smoking and obesity susceptibility loci on adolescent BMI: The National Longitudinal Study of Adolescent to Adult Health. BMC Genet 2015; 16:131. [PMID: 26537541 PMCID: PMC4634717 DOI: 10.1186/s12863-015-0289-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 10/29/2015] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Adolescence is a sensitive period for weight gain and risky health behaviors, such as smoking. Genome-wide association studies (GWAS) have identified loci contributing to adult body mass index (BMI). Evidence suggests that many of these loci have a larger influence on adolescent BMI. However, few studies have examined interactions between smoking and obesity susceptibility loci on BMI. This study investigates the interaction of current smoking and established BMI SNPs on adolescent BMI. Using data from the National Longitudinal Study of Adolescent to Adult Health, a nationally-representative, prospective cohort of the US school-based population in grades 7 to 12 (12-20 years of age) in 1994-95 who have been followed into adulthood (Wave II 1996; ages 12-21, Wave III; ages 18-27), we assessed (in 2014) interactions of 40 BMI-related SNPs and smoking status with percent of the CDC/NCHS 2000 median BMI (%MBMI) in European Americans (n = 5075), African Americans (n = 1744) and Hispanic Americans (n = 1294). RESULTS Two SNPs showed nominal significance for interaction (p < 0.05) between smoking and genotype with %MBMI in European Americans (EA) (rs2112347 (POC5): β = 1.98 (0.06, 3.90), p = 0.04 and near rs571312 (MC4R): β 2.15 (-0.03, 4.33) p = 0.05); and one SNP showed a significant interaction effect after stringent correction for multiple testing in Hispanic Americans (HA) (rs1514175 (TNNI3K): β 8.46 (4.32, 12.60), p = 5.9E-05). Stratifying by sex, these interactions suggest a stronger effect in female smokers. CONCLUSIONS Our study highlights potentially important sex differences in obesity risk by smoking status in adolescents, with those who may be most likely to initiate smoking (i.e., adolescent females), being at greatest risk for exacerbating genetic obesity susceptibility.
Collapse
Affiliation(s)
- Kristin L Young
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
- Carolina Population Center, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
- , 137 East Franklin Street, Suite 306, Chapel Hill, NC, 27514, USA.
| | - Misa Graff
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
- Carolina Population Center, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
- Carolina Center for Genome Sciences, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
| | - Andrea S Richardson
- Carolina Population Center, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
| | - Karen L Mohlke
- Carolina Center for Genome Sciences, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
- Department of Genetics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
| | - Leslie A Lange
- Carolina Center for Genome Sciences, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
- Department of Genetics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
| | - Ethan M Lange
- Carolina Center for Genome Sciences, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
- Department of Genetics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
| | - Kathleen M Harris
- Carolina Population Center, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
- Department of Sociology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
| | - Penny Gordon-Larsen
- Carolina Population Center, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC, USA.
| |
Collapse
|
41
|
Logan M, Van der Merwe MT, Dodgen TM, Myburgh R, Eloff A, Alessandrini M, Pepper MS. Allelic variants of the Melanocortin 4 receptor (MC4R) gene in a South African study group. Mol Genet Genomic Med 2015; 4:68-76. [PMID: 26788538 PMCID: PMC4707032 DOI: 10.1002/mgg3.180] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Revised: 09/09/2015] [Accepted: 09/11/2015] [Indexed: 12/20/2022] Open
Abstract
Obesity is a global epidemic that results in significant morbidity and mortality. Mutations in the melanocortin 4 receptor (MC4R) gene, which codes for a G-protein-coupled receptor responsible for postprandial satiety signaling, have been associated with monogenic obesity. The prevalence of obesity is on the increase in South Africa, and it is hypothesized that mutations in MC4R are a contributing factor. The aim of this study was to perform a retrospective assessment of the relationship between allelic variants of MC4R and BMI in a South African study cohort. DNA was isolated from a demographically representative cohort of 297 individuals and the entire MC4R gene sequenced by Sanger sequencing. Eight previously reported MC4R variants were identified in 42 of the 297 (14.1%) study participants. The most frequently observed MC4R alleles were V103I (4.0%), I170V (1.5%), and I198I (1.2%), while the remaining five variants together constituted 1.18%. Five compound heterozygotes were also detected. Although MC4R variants were rare, the majority of variation was observed in individuals of Black African ancestry. No statistically significant associations with BMI were reported. Given that lifestyle interventions have limited success in decreasing obesity, there is an urgent need to perform large-scale population studies to further elucidate the molecular underpinnings of this disease.
Collapse
Affiliation(s)
- Murray Logan
- Department of ImmunologyUniversity of PretoriaPretoriaSouth Africa; Faculty of Health SciencesInstitute for Cellular and Molecular MedicineUniversity of PretoriaPretoriaSouth Africa
| | | | - Tyren M Dodgen
- Faculty of Health SciencesInstitute for Cellular and Molecular MedicineUniversity of PretoriaPretoriaSouth Africa; Department of PharmacologyUniversity of PretoriaPretoriaSouth Africa
| | - Renier Myburgh
- Department of ImmunologyUniversity of PretoriaPretoriaSouth Africa; Faculty of Health SciencesInstitute for Cellular and Molecular MedicineUniversity of PretoriaPretoriaSouth Africa
| | - Arinda Eloff
- Department of ImmunologyUniversity of PretoriaPretoriaSouth Africa; Faculty of Health SciencesInstitute for Cellular and Molecular MedicineUniversity of PretoriaPretoriaSouth Africa
| | - Marco Alessandrini
- Department of ImmunologyUniversity of PretoriaPretoriaSouth Africa; Faculty of Health SciencesInstitute for Cellular and Molecular MedicineUniversity of PretoriaPretoriaSouth Africa
| | - Michael S Pepper
- Department of ImmunologyUniversity of PretoriaPretoriaSouth Africa; Faculty of Health SciencesInstitute for Cellular and Molecular MedicineUniversity of PretoriaPretoriaSouth Africa; Department of Genetic Medicine and DevelopmentFaculty of MedicineUniversity of GenevaGenevaSwitzerland
| |
Collapse
|
42
|
Fernandes AE, de Melo ME, Fujiwara CTH, Pioltine MB, Matioli SR, Santos A, Cercato C, Halpern A, Mancini MC. Associations between a common variant near the MC4R gene and serum triglyceride levels in an obese pediatric cohort. Endocrine 2015; 49:653-8. [PMID: 25948074 DOI: 10.1007/s12020-015-0616-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 04/21/2015] [Indexed: 11/25/2022]
Abstract
Polymorphisms near the MC4R gene may be related to an increased risk for obesity, but studies of variations in this gene and its relation to cardiometabolic profiles and food intake are scarce and controversial. The aim of this study is to evaluate the influence of the variants rs12970134 and rs17782313 near the MC4R gene in food intake, binge eating (BE) behavior, anthropometric parameters, body composition, metabolic profile, and cardiometabolic risk factors in obese children and adolescents. This is a cross-sectional study that included obese children and adolescents. We evaluated anthropometric, metabolic parameters and cardiometabolic risk factors, including hypertension, impaired fasting glucose, hypertriglyceridemia, and low HDL-cholesterol. BE was assessed through the BE scale, and a 24-h recall was used to evaluate total caloric intake and percentage of macronutrients and types of dietary fat. The MC4R variants rs12970134 and rs17782313 were genotyped using TaqMan assay. To assess the magnitude of risk, a logistic regression adjusted for Z-BMI, age, and gender was performed, adopting the significance level of 0.05. The study included 518 subjects (52.1 % girls, 12.7 ± 2.7 years old, Z-BMI = 3.24 ± 0.57). Carriers of the variant rs17782313 exhibit increased triglyceride levels (108 ± 48 vs. 119 ± 54, p = 0.034) and an increased risk of hypertriglyceridemia (OR 1.985, 95 % CI 1.288-3.057, p = 0.002). There was no association of the SNP rs12970134 with clinical, metabolic, or nutritional parameters. The variant rs12970134 and rs17782313 did not influence food intake or the presence of BE. The variant rs17782313 is associated with an increased risk of hypertriglyceridemia in obese children and adolescents.
Collapse
Affiliation(s)
- Ariana Ester Fernandes
- League of Childhood Obesity, Service of Endocrinology and Metabolism, Faculty of Medicine, Hospital das Clinicas, University of Sao Paulo, Av. Dr. Eneas de Carvalho Aguiar, 255, 7º andar, sala 7037, Sao Paulo, SP, 05403-000, Brazil,
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Micali N, Field AE, Treasure JL, Evans DM. Are obesity risk genes associated with binge eating in adolescence? Obesity (Silver Spring) 2015; 23:1729-36. [PMID: 26193063 PMCID: PMC4660437 DOI: 10.1002/oby.21147] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Revised: 04/02/2015] [Accepted: 04/08/2015] [Indexed: 12/20/2022]
Abstract
OBJECTIVE Cognitions and behaviors characteristic of binge eating are associated with a polymorphism in the FTO gene, robustly related to body mass index (BMI) and obesity risk. We investigated the association between binge eating and the individual and combined effect of 32 SNPs robustly associated with BMI in a population-based sample. We hypothesized that higher BMI and binge eating might share a common genetic etiology. METHODS Binge eating was assessed in adolescents from the Avon Longitudinal Study of Parents and Children at age 14 (n = 5,958) and 16 years (n = 4,948). We tested associations between 32 BMI-related SNPs and binge eating in crude and BMI-, age-, and gender-adjusted regression models. RESULTS Crude analyses showed an association between binge eating and rs1558902 (FTO) that persisted after adjustment for BMI (OR = 1.20, P = 8 × 10(-3) ). A weighted allelic score consisting of all 32 BMI-related SNPs was associated with binge eating (P = 8 × 10(-4) ); this association attenuated (P = 0.08) when rs1558902 was removed from the weighted allelic score. CONCLUSIONS BMI-related genes are associated with adolescent binge eating, in particular an FTO polymorphism. Although replication is needed, our findings have biological plausibility and are consistent with a postulated effect of FTO on appetite and food intake. Future studies should aim to understand the mechanisms underlying the relationship between FTO, binge eating, and obesity.
Collapse
Affiliation(s)
- Nadia Micali
- Population, Policy and Practice Research Programme, Child and Adolescent Mental Health Palliative Care and Pediatrics Section, Institute of Child Health, University College LondonLondon, UK
| | - Alison E Field
- Division of Adolescent Medicine, Department of Medicine, Boston Children's Hospital and Harvard Medical SchoolBoston, Massachusetts, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical SchoolBoston, Massachusetts, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public HealthBoston, Massachusetts, USA
| | - Janet L Treasure
- Section of Eating Disorders, Psychological Medicine, King's College London, Institute of PsychiatryLondon, UK
| | - David M Evans
- MRC Integrative Epidemiology Unit, University of BristolUK
- School of Social & Community Medicine, University of BristolBristol, UK
- University of Queensland Diamantina Institute, Translational Research InstituteBrisbane, Queensland, Australia
| |
Collapse
|
44
|
Gervasini G, Gamero-Villarroel C. Discussing the putative role of obesity-associated genes in the etiopathogenesis of eating disorders. Pharmacogenomics 2015; 16:1287-1305. [DOI: 10.2217/pgs.15.77] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
In addition to the identification of mutations clearly related to Mendelian forms of obesity; genome-wide association studies and follow-up studies have in the last years pinpointed several loci associated with BMI. These genetic alterations are located in or near genes expressed in the hypothalamus that are involved in the regulation of eating behavior. Accordingly, it seems plausible that these SNPs, or others located in related genes, could also help develop aberrant conduct patterns that favor the establishment of eating disorders should other susceptibility factors or personality dimensions be present. However, and somewhat surprisingly, with few exceptions such as BDNF, the great majority of the genes governing these pathways remain untested in patients with anorexia nervosa, bulimia nervosa or binge-eating disorder. In the present work, we review the few existing studies, but also indications and biological concepts that point to these genes in the CNS as good candidates for association studies with eating disorder patients.
Collapse
Affiliation(s)
- Guillermo Gervasini
- Department of Medical & Surgical Therapeutics, Division of Pharmacology, Medical School, University of Extremadura, Av. Elvas s/n, E-06005, Badajoz, Spain
| | - Carmen Gamero-Villarroel
- Department of Medical & Surgical Therapeutics, Division of Pharmacology, Medical School, University of Extremadura, Av. Elvas s/n, E-06005, Badajoz, Spain
| |
Collapse
|
45
|
Thompson MD, Hendy GN, Percy ME, Bichet DG, Cole DEC. G protein-coupled receptor mutations and human genetic disease. Methods Mol Biol 2015; 1175:153-87. [PMID: 25150870 DOI: 10.1007/978-1-4939-0956-8_8] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Genetic variations in G protein-coupled receptor genes (GPCRs) disrupt GPCR function in a wide variety of human genetic diseases. In vitro strategies and animal models have been used to identify the molecular pathologies underlying naturally occurring GPCR mutations. Inactive, overactive, or constitutively active receptors have been identified that result in pathology. These receptor variants may alter ligand binding, G protein coupling, receptor desensitization and receptor recycling. Receptor systems discussed include rhodopsin, thyrotropin, parathyroid hormone, melanocortin, follicle-stimulating hormone (FSH), luteinizing hormone, gonadotropin-releasing hormone (GNRHR), adrenocorticotropic hormone, vasopressin, endothelin-β, purinergic, and the G protein associated with asthma (GPRA or neuropeptide S receptor 1 (NPSR1)). The role of activating and inactivating calcium-sensing receptor (CaSR) mutations is discussed in detail with respect to familial hypocalciuric hypercalcemia (FHH) and autosomal dominant hypocalemia (ADH). The CASR mutations have been associated with epilepsy. Diseases caused by the genetic disruption of GPCR functions are discussed in the context of their potential to be selectively targeted by drugs that rescue altered receptors. Examples of drugs developed as a result of targeting GPCRs mutated in disease include: calcimimetics and calcilytics, therapeutics targeting melanocortin receptors in obesity, interventions that alter GNRHR loss from the cell surface in idiopathic hypogonadotropic hypogonadism and novel drugs that might rescue the P2RY12 receptor congenital bleeding phenotype. De-orphanization projects have identified novel disease-associated receptors, such as NPSR1 and GPR35. The identification of variants in these receptors provides genetic reagents useful in drug screens. Discussion of the variety of GPCRs that are disrupted in monogenic Mendelian disorders provides the basis for examining the significance of common pharmacogenetic variants.
Collapse
Affiliation(s)
- Miles D Thompson
- Department of Pharmacology, University of Toronto, 1 King's College Circle, Toronto, ON, Canada, M5S 1A8,
| | | | | | | | | |
Collapse
|
46
|
Abstract
Eating disorders (EDs) are serious psychiatric conditions influenced by biological, psychological, and sociocultural factors. A better understanding of the genetics of these complex traits and the development of more sophisticated molecular biology tools have advanced our understanding of the etiology of EDs. The aim of this review is to critically evaluate the literature on the genetic research conducted on three major EDs: anorexia nervosa (AN), bulimia nervosa (BN), and binge eating disorder (BED). We will first review the diagnostic criteria, clinical features, prevalence, and prognosis of AN, BN, and BED, followed by a review of family, twin, and adoption studies. We then review the history of genetic studies of EDs covering linkage analysis, candidate gene association studies, genome-wide association studies, and the study of rare variants in EDs. Our review also incorporates a translational perspective by covering animal models of ED-related phenotypes. Finally, we review the nascent field of epigenetics of EDs and a look forward to future directions for ED genetic research.
Collapse
Affiliation(s)
- Zeynep Yilmaz
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - J Andrew Hardaway
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Cynthia M Bulik
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
47
|
Role of ADHD symptoms as a contributing factor to obesity in patients with MC4R mutations. Med Hypotheses 2015; 84:4-7. [DOI: 10.1016/j.mehy.2014.11.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2014] [Revised: 10/27/2014] [Accepted: 11/06/2014] [Indexed: 02/04/2023]
|
48
|
Stijnen P, Tuand K, Varga TV, Franks PW, Aertgeerts B, Creemers JWM. The association of common variants in PCSK1 with obesity: a HuGE review and meta-analysis. Am J Epidemiol 2014; 180:1051-65. [PMID: 25355447 DOI: 10.1093/aje/kwu237] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Congenital deficiency of the proprotein convertase subtilisine/kexin type 1 gene (PCSK1), which encodes proprotein convertase 1/3, causes a severe multihormonal disorder marked by early-onset obesity. The single nucleotide polymorphisms (SNPs) rs6232 and rs6234-rs6235 in PCSK1 have been associated with obesity. However, case-control studies carried out in populations of different ethnicities have only partly replicated this association. Moreover, these SNPs have only weakly been associated with body mass index (weight (kg)/height (m)(2)) at a genome-wide level of significance. To investigate this discrepancy, we conducted a systematic search for studies published before December 2013 and extracted relevant data. Pooled estimates were calculated for overall and subgroup analyses. This meta-analysis confirmed the association of PCSK1 SNPs with obesity and provides the first evidence that the association between PCSK1 rs6232 and obesity is stronger for childhood obesity than for adult obesity. Moreover, we identified weak associations with body mass index and significantly stronger associations with waist circumference for rs6234-rs6235. No difference was found in the association with different obesity grades, and no association of PCSK1 rs6234-rs6235 with obesity was identified in Asian populations. This systematic Human Genome Epidemiology (HuGE) review showed convincingly that the SNPs rs6232, rs6234, and rs6235 in PCSK1 are associated with obesity in Caucasians.
Collapse
|
49
|
Common genetic variation in and near the melanocortin 4 receptor gene (MC4R) is associated with body mass index in American Indian adults and children. Hum Genet 2014; 133:1431-41. [PMID: 25103139 PMCID: PMC4185108 DOI: 10.1007/s00439-014-1477-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 08/01/2014] [Indexed: 01/30/2023]
Abstract
Six rare functional coding mutations were previously identified in melanocortin 4 receptor (MC4R) in 6,760 American Indians. Individuals heterozygous for one of these mutations become obese while young. We now investigate whether common non-coding variation near MC4R also contributes to obesity. Fifty-six tag single-nucleotide polymorphisms (SNPs) were genotyped in 3,229 full-heritage Pima Indians, and nine of these SNPs which showed evidence for association were genotyped in additional 3,852 mixed-heritage American Indians. Associations of SNPs with maximum body mass index (BMI) in adulthood (n = 5,918), BMI z score in childhood (n = 5,350), percent body fat (n = 864), energy expenditure (n = 358) and ad libitum food intake (n = 178) were assessed. Conditional analyses demonstrated that SNPs, rs74861148 and rs483125, were independently associated with BMI in adulthood (β = 0.68 kg/m2 per risk allele, p = 5 × 10−5; β = 0.58 kg/m2, p = 0.002, respectively) and BMI z score in childhood (β = 0.05, p = 0.02; β = 0.07, p = 0.01, respectively). One haplotype (frequency = 0.35) of the G allele at rs74861148 and the A allele at rs483125 provided the strongest evidence for association with adult BMI (β = 0.89 kg/m2, p = 5.5 × 10−7), and was also associated with childhood BMI z score (β = 0.08, p = 0.001). In addition, a promoter SNP rs11872992 was nominally associated with adult BMI (β = 0.61 kg/m2, p = 0.05) and childhood BMI z score (β = 0.11, p = 0.01), where the risk allele also modestly decreased transcription in vitro by 12 % (p = 0.005). This risk allele was further associated with increased percent body fat (β = 2.2 %, p = 0.002), increased food intake (β = 676 kcal/day, p = 0.007) and decreased energy expenditure (β = −53.4 kcal/day, p = 0.054). Common and rare variation in MC4R contributes to obesity in American Indians.
Collapse
|
50
|
Abstract
The past few decades have witnessed a rapid rise in nutrition-related disorders such as obesity in the United States and over the world. Traditional nutrition research has associated various foods and nutrients with obesity. Recent advances in genomics have led to identification of the genetic variants determining body weight and related dietary factors such as intakes of energy and macronutrients. In addition, compelling evidence has lent support to interactions between genetic variations and dietary factors in relation to obesity and weight change. Moreover, recently emerging data from other 'omics' studies such as epigenomics and metabolomics suggest that more complex interplays between the global features of human body and dietary factors may exist at multiple tiers in affecting individuals' susceptibility to obesity; and a concept of 'personalized nutrition' has been proposed to integrate this novel knowledge with traditional nutrition research, with the hope ultimately to endorse person-centric diet intervention to mitigate obesity and related disorders.
Collapse
Affiliation(s)
- Lu Qi
- Department of Nutrition, Harvard School of Public Health , Boston, Massachusetts , USA , and Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School , Boston, Massachusetts , USA
| |
Collapse
|