1
|
Li Y, Li J, Sun T, He Z, Liu C, Li Z, Wu Y, Xiang H. Sex-specific associations between body composition and depression among U.S. adults: a cross-sectional study. Lipids Health Dis 2025; 24:15. [PMID: 39827131 PMCID: PMC11742532 DOI: 10.1186/s12944-025-02437-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/11/2025] [Indexed: 01/22/2025] Open
Abstract
BACKGROUND Depression presents sexual dimorphism, and one important factor that increases the frequency of depression and contributes to sex-specific variations in its presentation is obesity. The conventional use of Body Mass Index (BMI) as an indicator of obesity is inherently limited due to its inability to distinguish between fat and lean mass, which limits its predictive utility for depression risk. Implementation of dual-energy X-ray absorptiometry (DXA) investigated sex-specific associations between body composition (fat mass, appendicular lean mass) and depression. METHODS Data from the NHANES cycles between 2011 and 2018 were analyzed, including 3,637 participants (1,788 males and 1,849 females). Four body composition profiles were identified in the subjects: low adiposity-low muscle (LA-LM), low adiposity-high muscle (LA-HM), high adiposity-low muscle (HA-LM) and high adiposity-high muscle (HA-HM). After accounting for confounding variables, the associations between fat mass index (FMI), appendicular skeletal muscle mass index (ASMI), body fat percentage (BFP), body composition phenotypes, and depression risk were assessed using restricted cubic spline (RCS) curves and multivariable logistic regression models. We further conducted interaction analyses for ASMI and FMI in females. RESULTS RCS curves indicated a U-shaped relationship between ASMI and the risk of depression in males. Logistic regression analysis revealed that in males, the second (OR = 0.43, 95%CI:0.22-0.85) and third (OR = 0.35, 95%CI:0.14-0.86) quartile levels of ASMI were significantly negatively associated with depression risk. In females, increases in BFP (OR = 1.06, 95%CI:1.03-1.09) and FMI (OR = 1.08, 95% CI:1.04-1.12) were significantly associated with an increased risk of depression. Additionally, compared to females with a low-fat high-muscle phenotype, those with LA-LM (OR = 3.97, 95%CI:2.16-7.30), HA-LM (OR = 5.40, 95%CI:2.34-12.46), and HA-HM (OR = 6.36, 95%CI:3.26-12.37) phenotypes were more likely to develop depression. Interestingly, further interaction analysis of ASMI and FMI in females revealed an interplay between height-adjusted fat mass and muscle mass (OR = 4.67, 95%CI: 2.04-10.71). CONCLUSION The findings demonstrate how important it is to consider body composition when estimating the risk of depression, particularly in females. There is a substantial correlation between the LA-LM, HA-LM, and HA-HM phenotypes in females with a higher prevalence of depression. It is advised to use a preventative approach that involves gaining muscle mass and losing fat.
Collapse
Affiliation(s)
- Yijing Li
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei, China
| | - Juan Li
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei, China
| | - Tianning Sun
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei, China
| | - Zhigang He
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei, China
| | - Cheng Liu
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei, China
| | - Zhixiao Li
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei, China
| | - Yanqiong Wu
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei, China.
| | - Hongbing Xiang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan, Hubei, China.
- Key Laboratory of Anesthesiology and Resuscitation , (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China.
| |
Collapse
|
2
|
Su F, Pfundstein G, Sah S, Zhang S, Keable R, Hagan DW, Sharpe LJ, Clemens KJ, Begg D, Phelps EA, Brown AJ, Leshchyns'ka I, Sytnyk V. Neuronal growth regulator 1 (NEGR1) promotes the synaptic targeting of glutamic acid decarboxylase 65 (GAD65). J Neurochem 2025; 169:e16279. [PMID: 39676071 DOI: 10.1111/jnc.16279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 11/15/2024] [Accepted: 11/20/2024] [Indexed: 12/17/2024]
Abstract
Neuronal growth regulator 1 (NEGR1) is a synaptic plasma membrane localized cell adhesion molecule implicated in a wide spectrum of psychiatric disorders. By RNAseq analysis of the transcriptomic changes in the brain of NEGR1-deficient mice, we found that NEGR1 deficiency affects the expression of the Gad2 gene. We show that glutamic acid decarboxylase 65 (GAD65), the Gad2 - encoded enzyme synthesizing the inhibitory neurotransmitter GABA on synaptic vesicles, accumulates non-synaptically in brains of NEGR1-deficient mice. The density of non-synaptic GAD65 accumulations is also increased in NEGR1 deficient cultured hypothalamic neurons, and this effect is rescued by re-expression of NEGR1. By using a novel biosensor of the plasma membrane attachment of GAD65, we demonstrate that GAD65 attaches to the plasma membrane. NEGR1 promotes palmitoylation-dependent clearance of GAD65 from the plasma membrane and targeting of GAD65 to plasma membrane-derived endocytic vesicles. In NEGR1 deficient cultured hypothalamic neurons, the synaptic and extrasynaptic levels of the plasma membrane attached GAD65 are increased, and the synaptic levels of GABA are reduced. NEGR1-deficient mice are characterized by reduced body weight, lower GABAergic synapse densities in the arcuate nucleus, and blunted responsiveness to the reinforcing effects of food rewards. Our results indicate that abnormalities in synaptic GABA synthesis can contribute to brain disorders associated with abnormal expression of NEGR1 in humans.
Collapse
Affiliation(s)
- Feifei Su
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Grant Pfundstein
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Saroj Sah
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Shuyue Zhang
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Ryan Keable
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - D Walker Hagan
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Laura J Sharpe
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Kelly J Clemens
- School of Psychology, The University of New South Wales, Sydney, New South Wales, Australia
| | - Denovan Begg
- School of Psychology, The University of New South Wales, Sydney, New South Wales, Australia
| | - Edward A Phelps
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, USA
| | - Andrew J Brown
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Iryna Leshchyns'ka
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| | - Vladimir Sytnyk
- School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
3
|
Streubel MK, Baumgartner A, Meier-Vollrath I, Frambach Y, Brandenburger M, Kisch T. Transcriptomics of Subcutaneous Tissue of Lipedema Identified Differentially Expressed Genes Involved in Adipogenesis, Inflammation, and Pain. PLASTIC AND RECONSTRUCTIVE SURGERY-GLOBAL OPEN 2024; 12:e6288. [PMID: 39525887 PMCID: PMC11548906 DOI: 10.1097/gox.0000000000006288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/16/2024] [Indexed: 11/16/2024]
Abstract
Background Lipedema is a disease typically affecting women with a symmetrical, painful fat distribution disorder, which is hypothesized to be caused by impaired adipogenesis, inflammation, and extracellular matrix remodeling, leading to fibrosis and the development of edema in lipedema subcutaneous adipose tissue. The pathogenesis and molecular processes leading to lipedema have not yet been clarified. Methods A whole transcriptome analysis of subcutaneous tissue of lipedema stages I (n = 12), II (n = 9), and III (n = 8) compared with hypertrophied subcutaneous tissue (n = 4) was performed. Further data about hormonal substitution and body morphology were collected. The study is registered at ClinicalTrials.gov (NCT05861583). Results We identified several differentially expressed genes involved in mechanisms leading to the development of lipedema. Some genes, such as PRKG2, MEDAG, CSF1R, BICC1, ERBB4, and ACP5, are involved in adipogenesis, regulating the development of mature adipocytes from mesenchymal stem cells. Other genes, such as MAFB, C1Q, C2, CD68, CD209, CD163, CD84, BCAT1, and TREM2, are predicted to be involved in lipid accumulation, hypertrophy, and the inflammation process. Further genes such as SHTN1, SCN7A, and SCL12A2 are predicted to be involved in the regulation and transmission of pain. Conclusions In summary, the pathogenesis and development of lipedema might be caused by alterations in adipogenesis, inflammation, and extracellular matrix remodeling, leading to fibrosis and the formation of edema resulting in this painful disease. These processes differ from hypertrophied adipose tissue and may therefore play a main role in the formation of lipedema.
Collapse
Affiliation(s)
- Maria Karolin Streubel
- From the University of Luebeck, Luebeck, Germany
- University Hospital Schleswig-Holstein, Luebeck, Germany
| | | | | | | | - Matthias Brandenburger
- Fraunhofer Research Institution for Individualized and Cell Based Medical Engineering, IMTE, Luebeck, Germany
| | - Tobias Kisch
- From the University of Luebeck, Luebeck, Germany
- Department of Plastic, Reconstructive and Aesthetic Surgery, Hand Surgery, Praxisklinik Kronshagen, Kiel-Kronshagen, Germany
| |
Collapse
|
4
|
Huang T, Zhang X, Li Q, Li X, Yao J, Song J, Chen Y, Ye L, Li C, Xiran P, Wen Y. The Association between Obesity Susceptibility and Polymorphisms of MC4R, SH2B1, and NEGR1 in Tibetans. Genet Test Mol Biomarkers 2024; 28:267-274. [PMID: 39034913 DOI: 10.1089/gtmb.2023.0546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024] Open
Abstract
Background: A high-altitude environment has inhibitory effects on obesity. Tibetans are not a high-risk population for obesity, but there are still obese individuals within that population. Obesity has become a worldwide health problem, and previous studies have found that obesity is closely associated with hereditary factors. Few studies have investigated obesity in Tibetans, and the association between gene polymorphisms and obesity in Tibetans remains unclear. Methods: Our study investigated the fat mass of 140 native Tibetan individuals (70 men and 70 women) from Lhasa and analyzed the associations between polymorphisms of melanocortin 4 receptor (MC4R), Src homology 2B adapter protein 1 (SH2B1), and neuronal growth regulator 1 (NEGR1) and obesity. Result: Among Tibetan individuals, there were differences in genotype and allele frequencies between those in the obesity group and those in the healthy group at MC4R (rs17782313) and SH2B1 (rs7359397). The polymorphisms of MC4R (rs17782313) were associated with fat mass and obesity in Tibetan men and women, and there was an association between SH2B1 (rs7359397) polymorphisms and fat mass and obesity in Tibetan men. However, polymorphisms of NEGR1 (rs3101336) were not associated with fat mass or obesity in Tibetan individuals. Conclusion: Among Tibetan individuals, polymorphisms of MC4R (rs17782313) and SH2B1 (rs7359397) were associated with obesity, but NEGR1 (rs3101336) polymorphisms were not associated with obesity.
Collapse
Affiliation(s)
- Ting Huang
- Institute of Biological Anthropology, Jinzhou Medical University, Jinzhou, China
| | - Xianpeng Zhang
- Institute of Biological Anthropology, Jinzhou Medical University, Jinzhou, China
| | - Qiang Li
- Institute of Biological Anthropology, Jinzhou Medical University, Jinzhou, China
- Department of Anatomy, Wuwei Occupational College, Wuwei, China
| | - Xin Li
- Institute of Biological Anthropology, Jinzhou Medical University, Jinzhou, China
| | - Jie Yao
- Institute of Biological Anthropology, Jinzhou Medical University, Jinzhou, China
| | - Jia Song
- Institute of Biological Anthropology, Jinzhou Medical University, Jinzhou, China
| | - Ying Chen
- Institute of Biological Anthropology, Jinzhou Medical University, Jinzhou, China
| | - Liping Ye
- Department of Radiology, Nagqu District People's Hospital, Nagqu, China
| | - Chunshan Li
- Department of Radiology, Nagqu District People's Hospital, Nagqu, China
| | - Pingcuo Xiran
- Maternal and Children Health Care Hospital of Ali District, Ali, China
| | - Youfeng Wen
- Institute of Biological Anthropology, Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
5
|
Littleton SH, Trang KB, Volpe CM, Cook K, DeBruyne N, Maguire JA, Weidekamp MA, Hodge KM, Boehm K, Lu S, Chesi A, Bradfield JP, Pippin JA, Anderson SA, Wells AD, Pahl MC, Grant SFA. Variant-to-function analysis of the childhood obesity chr12q13 locus implicates rs7132908 as a causal variant within the 3' UTR of FAIM2. CELL GENOMICS 2024; 4:100556. [PMID: 38697123 PMCID: PMC11099382 DOI: 10.1016/j.xgen.2024.100556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 03/21/2024] [Accepted: 04/08/2024] [Indexed: 05/04/2024]
Abstract
The ch12q13 locus is among the most significant childhood obesity loci identified in genome-wide association studies. This locus resides in a non-coding region within FAIM2; thus, the underlying causal variant(s) presumably influence disease susceptibility via cis-regulation. We implicated rs7132908 as a putative causal variant by leveraging our in-house 3D genomic data and public domain datasets. Using a luciferase reporter assay, we observed allele-specific cis-regulatory activity of the immediate region harboring rs7132908. We generated isogenic human embryonic stem cell lines homozygous for either rs7132908 allele to assess changes in gene expression and chromatin accessibility throughout a differentiation to hypothalamic neurons, a key cell type known to regulate feeding behavior. The rs7132908 obesity risk allele influenced expression of FAIM2 and other genes and decreased the proportion of neurons produced by differentiation. We have functionally validated rs7132908 as a causal obesity variant that temporally regulates nearby effector genes and influences neurodevelopment and survival.
Collapse
Affiliation(s)
- Sheridan H Littleton
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Khanh B Trang
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Christina M Volpe
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kieona Cook
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Psychiatry, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nicole DeBruyne
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jean Ann Maguire
- Center for Cellular and Molecular Therapeutics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Mary Ann Weidekamp
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Graduate Group in Genomics and Computational Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kenyaita M Hodge
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Keith Boehm
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Sumei Lu
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Alessandra Chesi
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jonathan P Bradfield
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Quantinuum Research LLC, San Diego, CA 92101, USA
| | - James A Pippin
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Stewart A Anderson
- Department of Child and Adolescent Psychiatry, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew D Wells
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew C Pahl
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Struan F A Grant
- Center for Spatial and Functional Genomics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Division of Human Genetics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Division of Endocrinology and Diabetes, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA.
| |
Collapse
|
6
|
Roumi Z, Salimi Z, Mahmoudi Z, Mobarakeh KA, Ladaninezhad M, Zeinalabedini M, Keshavarz Mohammadian M, Shamsi‐Goushki A, Saeedirad Z, Bahar B, Khoshdooz S, Kalantari N, Azizi Tabesh G, Doaei S, Gholamalizadeh M. Efficacy of a Comprehensive Weight Reduction Intervention in Male Adolescents With Different FTO Genotypes. Endocrinol Diabetes Metab 2024; 7:e00483. [PMID: 38556726 PMCID: PMC10982462 DOI: 10.1002/edm2.483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/29/2024] [Accepted: 03/07/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND The FTO gene polymorphisms may influence the effects of lifestyle interventions on obesity. The present study aimed to assess the influence of the rs9930506 FTO gene polymorphism on the success of a comprehensive weight loss intervention in male adolescents with overweight and obesity. METHODS This study was carried out on 96 adolescent boys with overweight and obesity who were randomly assigned to the intervention (n = 53) and control (n = 43) groups. The blood samples of the participants were collected, and the FTO gene was genotyped for the rs9930506 polymorphism. A comprehensive lifestyle intervention including changes in diet and physical activity was performed for 8 weeks in the intervention group. RESULTS Following the lifestyle intervention, BMI and fat mass decreased significantly in the intervention group compared with the control group (both p < 0.05), while no change was found in weight, height or body muscle percentage between the groups. The participants in the intervention group with the AA/AG genotype and not in carriers of the GG genotype had a significantly higher reduction in BMI (-1.21 vs. 1.87 kg/m2, F = 4.07, p < 0.05) compared with the control group. CONCLUSION The intervention in individuals with the AA/AG genotype has been significantly effective in weight loss compared with the control group. The intervention had no association effect on anthropometric indices in adolescents with the GG genotype of the FTO rs9930506 polymorphism. TRIAL REGISTRATION Name of the registry: National Nutrition and Food Technology Research Institute; Trial registration number: IRCT2016020925699N2; Date of registration: 24/04/2016; URL of trial registry record: https://www.irct.ir/trial/21447.
Collapse
Affiliation(s)
- Zahra Roumi
- Department of Nutrition, Science and Research BranchIslamic Azad UniversityTehranIran
| | - Zahra Salimi
- Nutrition and Metabolic Diseases Research CenterAhvaz Jundishapur University of Medical SciencesAhvazIran
| | - Zahra Mahmoudi
- Department of Nutrition, Science and Research BranchIslamic Azad UniversityTehranIran
| | - Khadijeh Abbasi Mobarakeh
- Food Security Research Center and Department of Community Nutrition, School of Nutrition and Food ScienceIsfahan University of Medical SciencesIsfahanIran
| | - Maryam Ladaninezhad
- School of Nutritional Sciences and DieteticsTehran University of Medical SciencesTehranIran
| | - Mobina Zeinalabedini
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and DieteticsTehran University of Medical SciencesTehranIran
| | | | - Ali Shamsi‐Goushki
- Department of Nutrition, School of MedicineMashhad University of Medical SciencesMashhadIran
| | - Zahra Saeedirad
- Department of Clinical Nutrition and DieteticsTehran University of Medical SciencesTehranIran
| | - Bojlul Bahar
- Nutrition Sciences and Applied Food Safety Studies, Research Centre for Global Development, School of Sport & Health SciencesUniversity of Central LancashirePrestonUK
| | - Sara Khoshdooz
- Faculty of MedicineGuilan University of Medical ScienceRashtIran
| | - Naser Kalantari
- Department of Community Nutrition, Faculty of Nutrition and Food TechnologyNational Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical SciencesTehranIran
| | - Ghasem Azizi Tabesh
- Genomic Research CenterShahid Beheshti University of Medical SciencesTehranIran
| | - Saeid Doaei
- Department of Community Nutrition, Faculty of Nutrition and Food TechnologyNational Nutrition and Food Technology Research Institute, Shahid Beheshti University of Medical SciencesTehranIran
| | | |
Collapse
|
7
|
Keller M, Svensson SIA, Rohde-Zimmermann K, Kovacs P, Böttcher Y. Genetics and Epigenetics in Obesity: What Do We Know so Far? Curr Obes Rep 2023; 12:482-501. [PMID: 37819541 DOI: 10.1007/s13679-023-00526-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/07/2023] [Indexed: 10/13/2023]
Abstract
PURPOSE OF REVIEW Enormous progress has been made in understanding the genetic architecture of obesity and the correlation of epigenetic marks with obesity and related traits. This review highlights current research and its challenges in genetics and epigenetics of obesity. RECENT FINDINGS Recent progress in genetics of polygenic traits, particularly represented by genome-wide association studies, led to the discovery of hundreds of genetic variants associated with obesity, which allows constructing polygenic risk scores (PGS). In addition, epigenome-wide association studies helped identifying novel targets and methylation sites being important in the pathophysiology of obesity and which are essential for the generation of methylation risk scores (MRS). Despite their great potential for predicting the individual risk for obesity, the use of PGS and MRS remains challenging. Future research will likely discover more loci being involved in obesity, which will contribute to better understanding of the complex etiology of human obesity. The ultimate goal from a clinical perspective will be generating highly robust and accurate prediction scores allowing clinicians to predict obesity as well as individual responses to body weight loss-specific life-style interventions.
Collapse
Affiliation(s)
- Maria Keller
- Medical Department III-Endocrinology, Nephrology, Rheumatology, Medical Center, University of Leipzig, 04103, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig, University Hospital Leipzig, 04103, Leipzig, Germany
| | - Stina Ingrid Alice Svensson
- EpiGen, Department of Clinical Molecular Biology, Institute of Clinical Medicine, University of Oslo, 0316, Oslo, Norway
| | - Kerstin Rohde-Zimmermann
- Medical Department III-Endocrinology, Nephrology, Rheumatology, Medical Center, University of Leipzig, 04103, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center Munich at the University of Leipzig, University Hospital Leipzig, 04103, Leipzig, Germany
| | - Peter Kovacs
- Medical Department III-Endocrinology, Nephrology, Rheumatology, Medical Center, University of Leipzig, 04103, Leipzig, Germany
| | - Yvonne Böttcher
- EpiGen, Department of Clinical Molecular Biology, Institute of Clinical Medicine, University of Oslo, 0316, Oslo, Norway.
- EpiGen, Medical Division, Akershus University Hospital, 1478, Lørenskog, Norway.
| |
Collapse
|
8
|
Salluzzo M, Vianello C, Abdullatef S, Rimondini R, Piccoli G, Carboni L. The Role of IgLON Cell Adhesion Molecules in Neurodegenerative Diseases. Genes (Basel) 2023; 14:1886. [PMID: 37895235 PMCID: PMC10606101 DOI: 10.3390/genes14101886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/26/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
In the brain, cell adhesion molecules (CAMs) are critical for neurite outgrowth, axonal fasciculation, neuronal survival and migration, and synapse formation and maintenance. Among CAMs, the IgLON family comprises five members: Opioid Binding Protein/Cell Adhesion Molecule Like (OPCML or OBCAM), Limbic System Associated Membrane Protein (LSAMP), neurotrimin (NTM), Neuronal Growth Regulator 1 (NEGR1), and IgLON5. IgLONs exhibit three N-terminal C2 immunoglobulin domains; several glycosylation sites; and a glycosylphosphatidylinositol anchoring to the membrane. Interactions as homo- or heterodimers in cis and in trans, as well as binding to other molecules, appear critical for their functions. Shedding by metalloproteases generates soluble factors interacting with cellular receptors and activating signal transduction. The aim of this review was to analyse the available data implicating a role for IgLONs in neuropsychiatric disorders. Starting from the identification of a pathological role for antibodies against IgLON5 in an autoimmune neurodegenerative disease with a poorly understood mechanism of action, accumulating evidence links IgLONs to neuropsychiatric disorders, albeit with still undefined mechanisms which will require future thorough investigations.
Collapse
Affiliation(s)
- Marco Salluzzo
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy;
| | - Clara Vianello
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (C.V.); (R.R.)
| | - Sandra Abdullatef
- Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy; (S.A.); (G.P.)
| | - Roberto Rimondini
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (C.V.); (R.R.)
| | - Giovanni Piccoli
- Department of Cellular, Computational and Integrative Biology, University of Trento, 38123 Trento, Italy; (S.A.); (G.P.)
| | - Lucia Carboni
- Department of Pharmacy and Biotechnology, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy;
| |
Collapse
|
9
|
Littleton SH, Trang KB, Volpe CM, Cook K, DeBruyne N, Ann Maguire J, Ann Weidekamp M, Boehm K, Chesi A, Pippin JA, Anderson SA, Wells AD, Pahl MC, Grant SF. Variant-to-function analysis of the childhood obesity chr12q13 locus implicates rs7132908 as a causal variant within the 3' UTR of FAIM2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.21.553157. [PMID: 37662342 PMCID: PMC10473629 DOI: 10.1101/2023.08.21.553157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
The ch12q13 obesity locus is among the most significant childhood obesity loci identified in genome-wide association studies. This locus resides in a non-coding region within FAIM2; thus, the underlying causal variant(s) presumably influence disease susceptibility via an influence on cis-regulation within the genomic region. We implicated rs7132908 as a putative causal variant at this locus leveraging a combination of our inhouse 3D genomic data, public domain datasets, and several computational approaches. Using a luciferase reporter assay in human primary astrocytes, we observed allele-specific cis-regulatory activity of the immediate region harboring rs7132908. Motivated by this finding, we went on to generate isogenic human embryonic stem cell lines homozygous for either rs7132908 allele with CRISPR-Cas9 homology-directed repair to assess changes in gene expression due to genotype and chromatin accessibility throughout a differentiation to hypothalamic neurons, a key cell type known to regulate feeding behavior. We observed that the rs7132908 obesity risk allele influenced the expression of FAIM2 along with other genes, decreased the proportion of neurons produced during differentiation, up-regulated cell death gene sets, and conversely down-regulated neuron differentiation gene sets. We have therefore functionally validated rs7132908 as a causal obesity variant which temporally regulates nearby effector genes at the ch12q13 locus and influences neurodevelopment and survival.
Collapse
Affiliation(s)
- Sheridan H. Littleton
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Khanh B. Trang
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Christina M. Volpe
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Biology, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kieona Cook
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Psychiatry, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Nicole DeBruyne
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jean Ann Maguire
- Center for Cellular and Molecular Therapeutics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Mary Ann Weidekamp
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Graduate Group in Genomics and Computational Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Keith Boehm
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Alessandra Chesi
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - James A. Pippin
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Stewart A. Anderson
- Department of Child and Adolescent Psychiatry, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Psychiatry, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andrew D. Wells
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Matthew C. Pahl
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Struan F.A. Grant
- Center for Spatial and Functional Genomics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Division of Human Genetics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Division of Endocrinology and Diabetes, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|
10
|
Novelli G, Cassadonte C, Sbraccia P, Biancolella M. Genetics: A Starting Point for the Prevention and the Treatment of Obesity. Nutrients 2023; 15:2782. [PMID: 37375686 DOI: 10.3390/nu15122782] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 06/15/2023] [Accepted: 06/16/2023] [Indexed: 06/29/2023] Open
Abstract
Obesity is a common, serious, and costly disease. More than 1 billion people worldwide are obese-650 million adults, 340 million adolescents, and 39 million children. The WHO estimates that, by 2025, approximately 167 million people-adults and children-will become less healthy because they are overweight or obese. Obesity-related conditions include heart disease, stroke, type 2 diabetes, and certain types of cancer. These are among the leading causes of preventable, premature death. The estimated annual medical cost of obesity in the United States was nearly $173 billion in 2019 dollars. Obesity is considered the result of a complex interaction between genes and the environment. Both genes and the environment change in different populations. In fact, the prevalence changes as the result of eating habits, lifestyle, and expression of genes coding for factors involved in the regulation of body weight, food intake, and satiety. Expression of these genes involves different epigenetic processes, such as DNA methylation, histone modification, or non-coding micro-RNA synthesis, as well as variations in the gene sequence, which results in functional alterations. Evolutionary and non-evolutionary (i.e., genetic drift, migration, and founder's effect) factors have shaped the genetic predisposition or protection from obesity in modern human populations. Understanding and knowing the pathogenesis of obesity will lead to prevention and treatment strategies not only for obesity, but also for other related diseases.
Collapse
Affiliation(s)
- Giuseppe Novelli
- Department of Biomedicine and Prevention, Medical School, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
- Italian Barometer Diabetes Observatory Foundation, IBDO, 00186 Rome, Italy
- Department of Pharmacology, School of Medicine, University of Nevada, Reno, NV 89557, USA
| | - Carmen Cassadonte
- Department of Biomedicine and Prevention, Medical School, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Paolo Sbraccia
- Italian Barometer Diabetes Observatory Foundation, IBDO, 00186 Rome, Italy
- Department of Systems Medicine, Medical School, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Michela Biancolella
- Department of Biology, Tor Vergata University of Rome, Via della Ricerca Scientifica 1, 00133 Rome, Italy
| |
Collapse
|
11
|
Wei Y, Han S, Wen J, Liao J, Liang J, Yu J, Chen X, Xiang S, Huang Z, Zhang B. E26 transformation-specific transcription variant 5 in development and cancer: modification, regulation and function. J Biomed Sci 2023; 30:17. [PMID: 36872348 PMCID: PMC9987099 DOI: 10.1186/s12929-023-00909-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 02/27/2023] [Indexed: 03/07/2023] Open
Abstract
E26 transformation-specific (ETS) transcription variant 5 (ETV5), also known as ETS-related molecule (ERM), exerts versatile functions in normal physiological processes, including branching morphogenesis, neural system development, fertility, embryonic development, immune regulation, and cell metabolism. In addition, ETV5 is repeatedly found to be overexpressed in multiple malignant tumors, where it is involved in cancer progression as an oncogenic transcription factor. Its roles in cancer metastasis, proliferation, oxidative stress response and drug resistance indicate that it is a potential prognostic biomarker, as well as a therapeutic target for cancer treatment. Post-translational modifications, gene fusion events, sophisticated cellular signaling crosstalk and non-coding RNAs contribute to the dysregulation and abnormal activities of ETV5. However, few studies to date systematically summarized the role and molecular mechanisms of ETV5 in benign diseases and in oncogenic progression. In this review, we specify the molecular structure and post-translational modifications of ETV5. In addition, its critical roles in benign and malignant diseases are summarized to draw a panorama for specialists and clinicians. The updated molecular mechanisms of ETV5 in cancer biology and tumor progression are delineated. Finally, we prospect the further direction of ETV5 research in oncology and its potential translational applications in the clinic.
Collapse
Affiliation(s)
- Yi Wei
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shenqi Han
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyuan Wen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyu Liao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Junnan Liang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingjing Yu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China
- Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Shuai Xiang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China.
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Zhao Huang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China.
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Bixiang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Clinical Medical Research Center of Hepatic Surgery at Hubei Province, Wuhan, China.
- Hubei Key Laboratory of Hepato-Pancreatic-Biliary Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, Wuhan, China.
- Key Laboratory of Organ Transplantation, National Health Commission, Wuhan, China.
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
12
|
Obesity-Related Genes Expression in Testes and Sperm Parameters Respond to GLP-1 and Caloric Restriction. Biomedicines 2022; 10:biomedicines10102609. [PMID: 36289871 PMCID: PMC9599882 DOI: 10.3390/biomedicines10102609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/27/2022] [Accepted: 10/04/2022] [Indexed: 11/25/2022] Open
Abstract
Aim: Calorie restriction (CR) diets and glucagon-Like Peptide-1 (GLP-1) analogs are known to alter energy homeostasis with the potential to affect the expression of obesity-related genes (ORGs). We hypothesized that CR and GLP-1 administration can alter ORGs expression in spermatozoa and testes, as well as the sperm parameters implicated in male fertility. Materials and Methods: Six-week-old adult male Wistar rats (n = 16) were divided into three groups, submitted either to CR (n = 6, fed with 30% less chow diet than the control rats), GLP-1 administration (n = 5, 3.5 pmol/min/kg intraperitoneal) for 28 days, or used as controls (n = 5, fed ad libitum). Selected ORGs expression, namely the fat mass and obesity-associated (FTO), melanocortin-4 receptor (MC4R), glucosamine-6-phosphate deaminase 2 (GNPDA2), and transmembrane protein 18 (TMEM18) were evaluated in testes and spermatozoa by a quantitative polymerase chain reaction (qPCR). Results: CR resulted in lower body weight gain and insulin resistance, but a higher percentage of sperm head defects. GLP-1 administration, despite showing no influence on body weight or glucose homeostasis, resulted in a lower percentage of sperm head defects. CR and GLP-1 administration were associated with a higher expression of all ORGs in the testes. Under CR conditions, the genes FTO and TMEM18 expression in the testes and the MC4R and TMEM18 transcripts abundance in sperm were positively correlated with the spermatozoa oxidative status. The abundance of FTO and TMEM18 in the spermatozoa of rats under CR were positively correlated with sperm concentration, while the testes’ TMEM18 expression was also positively correlated with sperm vitality and negatively correlated with insulin resistance. Testes GNPDA2 expression was negatively correlated with sperm head defects. Conclusions: CR and GLP-1 administration results in higher ORGs expression in testes, and these were correlated with several alterations in sperm fertility parameters.
Collapse
|
13
|
Takeuchi H, Jimbo H, Sumiyoshi A, Omori A, Nakane K, Tabuchi E. Differential Effect of Fat Intake on Blood Pressure in Healthy Japanese Women with and without the Variant of Fat Mass and Obesity-Associated Gene. J Oleo Sci 2022; 71:1511-1519. [PMID: 36089400 DOI: 10.5650/jos.ess22153] [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: 11/13/2022] Open
Abstract
A high-fat diet is believed to be a risk factor for hypertension through inducing obesity. It has been reported that variants of the fat mass and obesity-associated (FTO) and beta-3 adrenergic receptor (B3AR) genes are associated with obesity and blood pressure. The purpose of this study was to investigate the effect of dietary fat on blood pressure with or without the variant of the FTO and B3AR genes. A total of 227 healthy Japanese women aged 18 to 64 years were recruited for measurement of nutrient intake and blood pressure. The single nucleotide polymorphism rs9939609 of the FTO gene and rs4994 of the B3AR gene were genotyped. Spearman's rank correlation coefficient was applied to investigate the relationship between fat intake and blood pressure. A hierarchical multiple regression analysis was performed to determine whether the genotype interacts with fat intake to affect blood pressure. No significant correlations were found between fat intake and either systolic or diastolic blood pressure. A significant negative correlation was found between fat intake and both blood pressures in the FTO-gene-variant group, but not in the normal-FTO-gene group. In hierarchical multiple regression analysis, the interaction of fat intake and the gene variant showed significance, and the change in coefficient of determination (R 2) was significantly increased with increases of the interaction variable. These results indicate that the effect of fat intake on blood pressure may be modified by the variant of the FTO gene such that a high-fat diet intake may be associated with a decrease of systolic and diastolic blood pressure in healthy Japanese women with the FTO variant. Our results did not support the hypothesis that a high-fat diet increases blood pressure.
Collapse
Affiliation(s)
| | - Hiroko Jimbo
- Department of Food and Nutrition, Toyama College
| | - Ai Sumiyoshi
- Department of Food and Nutrition, Toyama College
| | - Akira Omori
- Department of Food and Nutrition, Toyama College
| | - Kazue Nakane
- Department of Food and Nutrition, Toyama College
| | | |
Collapse
|
14
|
Leyden GM, Shapland CY, Davey Smith G, Sanderson E, Greenwood MP, Murphy D, Richardson TG. Harnessing tissue-specific genetic variation to dissect putative causal pathways between body mass index and cardiometabolic phenotypes. Am J Hum Genet 2022; 109:240-252. [PMID: 35090585 PMCID: PMC8874216 DOI: 10.1016/j.ajhg.2021.12.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/14/2021] [Indexed: 12/11/2022] Open
Abstract
Body mass index (BMI) is a complex disease risk factor known to be influenced by genes acting via both metabolic pathways and appetite regulation. In this study, we aimed to gain insight into the phenotypic consequences of BMI-associated genetic variants, which may be mediated by their expression in different tissues. First, we harnessed meta-analyzed gene expression datasets derived from subcutaneous adipose (n = 1257) and brain (n = 1194) tissue to identify 86 and 140 loci, respectively, which provided evidence of genetic colocalization with BMI. These two sets of tissue-partitioned loci had differential effects with respect to waist-to-hip ratio, suggesting that the way they influence fat distribution might vary despite their having very similar average magnitudes of effect on BMI itself (adipose = 0.0148 and brain = 0.0149 standard deviation change in BMI per effect allele). For instance, BMI-associated variants colocalized with TBX15 expression in adipose tissue (posterior probability [PPA] = 0.97), but not when we used TBX15 expression data derived from brain tissue (PPA = 0.04) This gene putatively influences BMI via its role in skeletal development. Conversely, there were loci where BMI-associated variants provided evidence of colocalization with gene expression in brain tissue (e.g., NEGR1, PPA = 0.93), but not when we used data derived from adipose tissue, suggesting that these genes might be more likely to influence BMI via energy balance. Leveraging these tissue-partitioned variant sets through a multivariable Mendelian randomization framework provided strong evidence that the brain-tissue-derived variants are predominantly responsible for driving the genetically predicted effects of BMI on cardiovascular-disease endpoints (e.g., coronary artery disease: odds ratio = 1.05, 95% confidence interval = 1.04-1.07, p = 4.67 × 10-14). In contrast, our analyses suggested that the adipose tissue variants might predominantly be responsible for the underlying relationship between BMI and measures of cardiac function, such as left ventricular stroke volume (beta = 0.21, 95% confidence interval = 0.09-0.32, p = 6.43 × 10-4).
Collapse
Affiliation(s)
- Genevieve M Leyden
- MRC Integrative Epidemiology Unit, Bristol Population Health Science Institute, University of Bristol, Bristol, BS8 2BN, United Kingdom; Bristol Medical School: Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, United Kingdom.
| | - Chin Yang Shapland
- MRC Integrative Epidemiology Unit, Bristol Population Health Science Institute, University of Bristol, Bristol, BS8 2BN, United Kingdom
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, Bristol Population Health Science Institute, University of Bristol, Bristol, BS8 2BN, United Kingdom
| | - Eleanor Sanderson
- MRC Integrative Epidemiology Unit, Bristol Population Health Science Institute, University of Bristol, Bristol, BS8 2BN, United Kingdom
| | - Michael P Greenwood
- Bristol Medical School: Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, United Kingdom
| | - David Murphy
- Bristol Medical School: Translational Health Sciences, Dorothy Hodgkin Building, University of Bristol, Bristol, BS1 3NY, United Kingdom
| | - Tom G Richardson
- MRC Integrative Epidemiology Unit, Bristol Population Health Science Institute, University of Bristol, Bristol, BS8 2BN, United Kingdom; Novo Nordisk Research Centre, Headington, Oxford, OX3 7FZ, United Kingdom.
| |
Collapse
|
15
|
Plaza-Florido A, Pérez-Prieto I, Molina-Garcia P, Radom-Aizik S, Ortega FB, Altmäe S. Transcriptional and Epigenetic Response to Sedentary Behavior and Physical Activity in Children and Adolescents: A Systematic Review. Front Pediatr 2022; 10:917152. [PMID: 35813370 PMCID: PMC9263076 DOI: 10.3389/fped.2022.917152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 06/02/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The links of sedentary behavior and physical activity with health outcomes in children and adolescents is well known. However, the molecular mechanisms involved are poorly understood. We aimed to synthesize the current knowledge of the association of sedentary behavior and physical activity (acute and chronic effects) with gene expression and epigenetic modifications in children and adolescents. METHODS PubMed, Web of Science, and Scopus databases were systematically searched until April 2022. A total of 15 articles were eligible for this review. The risk of bias assessment was performed using the Joanna Briggs Institute Critical Appraisal Tool for Systematic Reviews and/or a modified version of the Downs and Black checklist. RESULTS Thirteen studies used candidate gene approach, while only 2 studies performed high-throughput analyses. The candidate genes significantly linked to sedentary behavior or physical activity were: FOXP3, HSD11B2, IL-10, TNF-α, ADRB2, VEGF, HSP70, SOX, and GPX. Non-coding Ribonucleic acids (RNAs) regulated by sedentary behavior or physical activity were: miRNA-222, miRNA-146a, miRNA-16, miRNA-126, miR-320a, and long non-coding RNA MALAT1. These molecules are involved in inflammation, immune function, angiogenic process, and cardiovascular disease. Transcriptomics analyses detected thousands of genes that were altered following an acute bout of physical activity and are linked to gene pathways related to immune function, apoptosis, and metabolic diseases. CONCLUSION The evidence found to date is rather limited. Multidisciplinary studies are essential to characterize the molecular mechanisms in response to sedentary behavior and physical activity in the pediatric population. Larger cohorts and randomized controlled trials, in combination with multi-omics analyses, may provide the necessary data to bring the field forward. SYSTEMATIC REVIEW REGISTRATION [www.ClinicalTrials.gov], identifier [CRD42021235431].
Collapse
Affiliation(s)
- Abel Plaza-Florido
- Department of Physical and Sports Education, Faculty of Sport Sciences, PROFITH "PROmoting FITness and Health Through Physical Activity" Research Group, Sport and Health University Research Institute (iMUDS), University of Granada, Granada, Spain
| | - Inmaculada Pérez-Prieto
- Department of Biochemistry and Molecular Biology, Faculty of Sciences, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria (ibs.GRANADA), Granada, Spain
| | - Pablo Molina-Garcia
- Department of Physical and Sports Education, Faculty of Sport Sciences, PROFITH "PROmoting FITness and Health Through Physical Activity" Research Group, Sport and Health University Research Institute (iMUDS), University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria (ibs.Granada), Physical Medicine and Rehabilitation Service, Virgen de las Nieves University Hospital, Granada, Spain
| | - Shlomit Radom-Aizik
- Pediatric Exercise and Genomics Research Center, UC Irvine School of Medicine, Irvine, CA, United States
| | - Francisco B Ortega
- Department of Physical and Sports Education, Faculty of Sport Sciences, PROFITH "PROmoting FITness and Health Through Physical Activity" Research Group, Sport and Health University Research Institute (iMUDS), University of Granada, Granada, Spain.,Faculty of Sport and Health Sciences, University of Jyväskylä, Jyväskylä, Finland.,Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Signe Altmäe
- Department of Biochemistry and Molecular Biology, Faculty of Sciences, University of Granada, Granada, Spain.,Instituto de Investigación Biosanitaria (ibs.GRANADA), Granada, Spain.,Division of Obstetrics and Gynecology, CLINTEC, Karolinska Institutet, Stockholm, Sweden.,Competence Centre on Health Technologies, Tartu, Estonia
| |
Collapse
|
16
|
Szentkereszty-Kovács Z, Fiatal S, Janka EA, Kovács D, Szegedi A, Remenyik É, Törőcsik D. Leptin Receptor (rs1137101) and Brain-Derived Neurotrophic Factor (rs925946) Gene Variants Are Associated with Obesity in the Early- but Not in the Late-Onset Population of Hungarian Psoriatic Patients. Life (Basel) 2021; 11:life11101086. [PMID: 34685457 PMCID: PMC8538564 DOI: 10.3390/life11101086] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/09/2021] [Accepted: 10/12/2021] [Indexed: 11/16/2022] Open
Abstract
Background: Psoriatic patients have considerably higher odds of being obese compared with the general population; however, the exact pathophysiological link between psoriasis and obesity needs to be elucidated. Methods: To investigate the association of psoriasis with established obesity-related gene variants, we conducted a population-based case-control study including 3541 subjects (574 psoriasis cases and 2967 controls from the general Hungarian population). Genotyping of 20 SNPs at ADIPOQ, BDNF, FTO, GNPDA2, LEPR, MC4R, NEGR1, NPY, PPARG, TMEM18, and UCP2 were determined, and differences in genotype and allele distributions were investigated. Multiple logistic regression analyses were implemented. Results: Analysis revealed an association between the G allele of the rs1137101 polymorphism (LEPR gene) and obesity risk (OR: 3.30 (1.45; 7.50), p = 0.004) in the early-onset group of psoriatic patients. Furthermore, the T allele of rs925946 polymorphism (BDNF gene) was also associated with increased risk of obesity in early-onset psoriasis (OR: 2.26 (1.24; 4.14), p = 0.008). Conclusions: Our results suggest that in psoriatic patients, there are prominent differences in the causes of obesity that should be accounted for, including not only environmental factors but also patient characteristics, such as the time of disease onset as well as genetic factors.
Collapse
Affiliation(s)
- Zita Szentkereszty-Kovács
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98, 4032 Debrecen, Hungary; (Z.S.-K.); (E.A.J.); (D.K.); (A.S.); (É.R.)
| | - Szilvia Fiatal
- Department of Public Health and Epidemiology, Faculty of Medicine, University of Debrecen, Kassai út 26, 4012 Debrecen, Hungary;
| | - Eszter Anna Janka
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98, 4032 Debrecen, Hungary; (Z.S.-K.); (E.A.J.); (D.K.); (A.S.); (É.R.)
| | - Dóra Kovács
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98, 4032 Debrecen, Hungary; (Z.S.-K.); (E.A.J.); (D.K.); (A.S.); (É.R.)
| | - Andrea Szegedi
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98, 4032 Debrecen, Hungary; (Z.S.-K.); (E.A.J.); (D.K.); (A.S.); (É.R.)
| | - Éva Remenyik
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98, 4032 Debrecen, Hungary; (Z.S.-K.); (E.A.J.); (D.K.); (A.S.); (É.R.)
| | - Dániel Törőcsik
- Department of Dermatology, Faculty of Medicine, University of Debrecen, Nagyerdei Krt. 98, 4032 Debrecen, Hungary; (Z.S.-K.); (E.A.J.); (D.K.); (A.S.); (É.R.)
- Correspondence: ; Tel.: +36-52-255-602
| |
Collapse
|
17
|
Kaare M, Mikheim K, Lilleväli K, Kilk K, Jagomäe T, Leidmaa E, Piirsalu M, Porosk R, Singh K, Reimets R, Taalberg E, Schäfer MKE, Plaas M, Vasar E, Philips MA. High-Fat Diet Induces Pre-Diabetes and Distinct Sex-Specific Metabolic Alterations in Negr1-Deficient Mice. Biomedicines 2021; 9:1148. [PMID: 34572334 PMCID: PMC8466019 DOI: 10.3390/biomedicines9091148] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/27/2021] [Accepted: 08/28/2021] [Indexed: 11/16/2022] Open
Abstract
In the large GWAS studies, NEGR1 gene has been one of the most significant gene loci for body mass phenotype. The purpose of the current study was to clarify the role of NEGR1 in the maintenance of systemic metabolism, including glucose homeostasis, by using both male and female Negr1-/- mice receiving a standard or high fat diet (HFD). We found that 6 weeks of HFD leads to higher levels of blood glucose in Negr1-/- mice. In the glucose tolerance test, HFD induced phenotype difference only in male mice; Negr1-/- male mice displayed altered glucose tolerance, accompanied with upregulation of circulatory branched-chain amino acids (BCAA). The general metabolomic profile indicates that Negr1-/- mice are biased towards glyconeogenesis, fatty acid synthesis, and higher protein catabolism, all of which are amplified by HFD. Negr1 deficiency appears to induce alterations in the efficiency of energy storage; reduced food intake could be an attempt to compensate for the metabolic challenge present in the Negr1-/- males, particularly during the HFD exposure. Our results suggest that the presence of functional Negr1 allows male mice to consume more HFD and prevents the development of glucose intolerance, liver steatosis, and excessive weight gain.
Collapse
Affiliation(s)
- Maria Kaare
- Institute of Biomedicine and Translational Medicine, Department of Physiology, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (K.M.); (K.L.); (T.J.); (M.P.); (K.S.); (E.V.); (M.-A.P.)
- Center of Excellence in Genomics and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; (K.K.); (R.P.); (E.T.)
| | - Kaie Mikheim
- Institute of Biomedicine and Translational Medicine, Department of Physiology, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (K.M.); (K.L.); (T.J.); (M.P.); (K.S.); (E.V.); (M.-A.P.)
- Center of Excellence in Genomics and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; (K.K.); (R.P.); (E.T.)
| | - Kersti Lilleväli
- Institute of Biomedicine and Translational Medicine, Department of Physiology, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (K.M.); (K.L.); (T.J.); (M.P.); (K.S.); (E.V.); (M.-A.P.)
- Center of Excellence in Genomics and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; (K.K.); (R.P.); (E.T.)
| | - Kalle Kilk
- Center of Excellence in Genomics and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; (K.K.); (R.P.); (E.T.)
- Institute of Biomedicine and Translational Medicine, Department of Biochemistry, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia
| | - Toomas Jagomäe
- Institute of Biomedicine and Translational Medicine, Department of Physiology, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (K.M.); (K.L.); (T.J.); (M.P.); (K.S.); (E.V.); (M.-A.P.)
- Center of Excellence in Genomics and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; (K.K.); (R.P.); (E.T.)
- Institute of Biomedicine and Translational Medicine, Laboratory Animal Center, University of Tartu, 14B Ravila Street, 50411 Tartu, Estonia; (R.R.); (M.P.)
| | - Este Leidmaa
- Institute of Molecular Psychiatry, Medical Faculty, University of Bonn, 53129 Bonn, Germany;
| | - Maria Piirsalu
- Institute of Biomedicine and Translational Medicine, Department of Physiology, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (K.M.); (K.L.); (T.J.); (M.P.); (K.S.); (E.V.); (M.-A.P.)
- Center of Excellence in Genomics and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; (K.K.); (R.P.); (E.T.)
| | - Rando Porosk
- Center of Excellence in Genomics and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; (K.K.); (R.P.); (E.T.)
- Institute of Biomedicine and Translational Medicine, Department of Biochemistry, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia
| | - Katyayani Singh
- Institute of Biomedicine and Translational Medicine, Department of Physiology, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (K.M.); (K.L.); (T.J.); (M.P.); (K.S.); (E.V.); (M.-A.P.)
- Center of Excellence in Genomics and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; (K.K.); (R.P.); (E.T.)
| | - Riin Reimets
- Institute of Biomedicine and Translational Medicine, Laboratory Animal Center, University of Tartu, 14B Ravila Street, 50411 Tartu, Estonia; (R.R.); (M.P.)
| | - Egon Taalberg
- Center of Excellence in Genomics and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; (K.K.); (R.P.); (E.T.)
- Institute of Biomedicine and Translational Medicine, Department of Biochemistry, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia
| | - Michael K. E. Schäfer
- Department of Anesthesiology, Focus Program Translational Neurosciences, Research Center for Immunotherapy, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany;
| | - Mario Plaas
- Institute of Biomedicine and Translational Medicine, Laboratory Animal Center, University of Tartu, 14B Ravila Street, 50411 Tartu, Estonia; (R.R.); (M.P.)
| | - Eero Vasar
- Institute of Biomedicine and Translational Medicine, Department of Physiology, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (K.M.); (K.L.); (T.J.); (M.P.); (K.S.); (E.V.); (M.-A.P.)
- Center of Excellence in Genomics and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; (K.K.); (R.P.); (E.T.)
| | - Mari-Anne Philips
- Institute of Biomedicine and Translational Medicine, Department of Physiology, University of Tartu, 19 Ravila Street, 50411 Tartu, Estonia; (K.M.); (K.L.); (T.J.); (M.P.); (K.S.); (E.V.); (M.-A.P.)
- Center of Excellence in Genomics and Translational Medicine, University of Tartu, 50411 Tartu, Estonia; (K.K.); (R.P.); (E.T.)
| |
Collapse
|
18
|
Ren J, Wu NN, Wang S, Sowers JR, Zhang Y. Obesity cardiomyopathy: evidence, mechanisms, and therapeutic implications. Physiol Rev 2021; 101:1745-1807. [PMID: 33949876 PMCID: PMC8422427 DOI: 10.1152/physrev.00030.2020] [Citation(s) in RCA: 215] [Impact Index Per Article: 53.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The prevalence of heart failure is on the rise and imposes a major health threat, in part, due to the rapidly increased prevalence of overweight and obesity. To this point, epidemiological, clinical, and experimental evidence supports the existence of a unique disease entity termed “obesity cardiomyopathy,” which develops independent of hypertension, coronary heart disease, and other heart diseases. Our contemporary review evaluates the evidence for this pathological condition, examines putative responsible mechanisms, and discusses therapeutic options for this disorder. Clinical findings have consolidated the presence of left ventricular dysfunction in obesity. Experimental investigations have uncovered pathophysiological changes in myocardial structure and function in genetically predisposed and diet-induced obesity. Indeed, contemporary evidence consolidates a wide array of cellular and molecular mechanisms underlying the etiology of obesity cardiomyopathy including adipose tissue dysfunction, systemic inflammation, metabolic disturbances (insulin resistance, abnormal glucose transport, spillover of free fatty acids, lipotoxicity, and amino acid derangement), altered intracellular especially mitochondrial Ca2+ homeostasis, oxidative stress, autophagy/mitophagy defect, myocardial fibrosis, dampened coronary flow reserve, coronary microvascular disease (microangiopathy), and endothelial impairment. Given the important role of obesity in the increased risk of heart failure, especially that with preserved systolic function and the recent rises in COVID-19-associated cardiovascular mortality, this review should provide compelling evidence for the presence of obesity cardiomyopathy, independent of various comorbid conditions, underlying mechanisms, and offer new insights into potential therapeutic approaches (pharmacological and lifestyle modification) for the clinical management of obesity cardiomyopathy.
Collapse
Affiliation(s)
- Jun Ren
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China.,Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington
| | - Ne N Wu
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
| | - Shuyi Wang
- School of Medicine, Shanghai University, Shanghai, China.,University of Wyoming College of Health Sciences, Laramie, Wyoming
| | - James R Sowers
- Dalton Cardiovascular Research Center, Diabetes and Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - Yingmei Zhang
- Department of Cardiology, Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital Fudan University, Shanghai, China
| |
Collapse
|
19
|
Marderstein AR, Davenport ER, Kulm S, Van Hout CV, Elemento O, Clark AG. Leveraging phenotypic variability to identify genetic interactions in human phenotypes. Am J Hum Genet 2021; 108:49-67. [PMID: 33326753 PMCID: PMC7820920 DOI: 10.1016/j.ajhg.2020.11.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 11/23/2020] [Indexed: 12/13/2022] Open
Abstract
Although thousands of loci have been associated with human phenotypes, the role of gene-environment (GxE) interactions in determining individual risk of human diseases remains unclear. This is partly because of the severe erosion of statistical power resulting from the massive number of statistical tests required to detect such interactions. Here, we focus on improving the power of GxE tests by developing a statistical framework for assessing quantitative trait loci (QTLs) associated with the trait means and/or trait variances. When applying this framework to body mass index (BMI), we find that GxE discovery and replication rates are significantly higher when prioritizing genetic variants associated with the variance of the phenotype (vQTLs) compared to when assessing all genetic variants. Moreover, we find that vQTLs are enriched for associations with other non-BMI phenotypes having strong environmental influences, such as diabetes or ulcerative colitis. We show that GxE effects first identified in quantitative traits such as BMI can be used for GxE discovery in disease phenotypes such as diabetes. A clear conclusion is that strong GxE interactions mediate the genetic contribution to body weight and diabetes risk.
Collapse
Affiliation(s)
- Andrew R Marderstein
- Tri-Institutional Program in Computational Biology & Medicine, Weill Cornell Medicine, New York, NY 10021, USA; Institute of Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA; Department of Computational Biology, Cornell University, Ithaca, NY 14850, USA
| | - Emily R Davenport
- Department of Biology, Huck Institutes of the Life Sciences, Institute for Computational and Data Sciences, Pennsylvania State University, University Park, PA 16802, USA
| | - Scott Kulm
- Institute of Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | | | - Olivier Elemento
- Institute of Computational Biomedicine, Weill Cornell Medicine, New York, NY 10021, USA; Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA.
| | - Andrew G Clark
- Department of Computational Biology, Cornell University, Ithaca, NY 14850, USA.
| |
Collapse
|
20
|
Mao Z, Feng M, Li Z, Zhou M, Xu L, Pan K, Wang S, Su W, Zhang W. ETV5 Regulates Hepatic Fatty Acid Metabolism Through PPAR Signaling Pathway. Diabetes 2021; 70:214-226. [PMID: 33093014 DOI: 10.2337/db20-0619] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 10/15/2020] [Indexed: 11/13/2022]
Abstract
ETV5 is an ETS transcription factor that has been associated with obesity in genomic association studies. However, little is known about the role of ETV5 in hepatic lipid metabolism and nonalcoholic fatty liver disease. In the current study, we found that ETV5 protein expression was increased in diet- and genetically induced steatotic liver. ETV5 responded to the nutrient status in a mammalian target of rapamycin complex 1 (mTORC1)-dependent manner and in turn, regulated mTORC1 activity. Both viral-mediated and genetic depletion of ETV5 in mice led to increased lipid accumulation in the liver. RNA sequencing analysis revealed that peroxisome proliferator-activated receptor (PPAR) signaling and fatty acid degradation/metabolism pathways were significantly downregulated in ETV5-deficient hepatocytes in vivo and in vitro. Mechanistically, ETV5 could bind to the PPAR response element region of downstream genes and enhance its transactivity. Collectively, our study identifies ETV5 as a novel transcription factor for the regulation of hepatic fatty acid metabolism, which is required for the optimal β-oxidation process. ETV5 may provide a therapeutic target for the treatment of hepatic steatosis.
Collapse
Affiliation(s)
- Zhuo Mao
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong Province, China
| | - Mingji Feng
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong Province, China
| | - Zhuoran Li
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong Province, China
| | - Minsi Zhou
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong Province, China
| | - Langning Xu
- Department of Neurosurgery, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, China
| | - Ke Pan
- Institute for Advanced Study, Shenzhen University, Shenzhen, Guangdong Province, China
| | - Shaoxiang Wang
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong Province, China
| | - Wen Su
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong Province, China
| | - Weizhen Zhang
- Department of Physiology and Pathophysiology, School of Basic Science, Peking University Health Science Center, Beijing, China
| |
Collapse
|
21
|
Abstract
The pathophysiology of obesity is complex and includes changes in eating behavior, genetic, epigenetic, environmental factors, and much more. To date, ~40 genetic polymorphisms are associated with obesity and fat distribution. However, since these options do not fully explain the inheritance of obesity, other options, such as epigenetic changes, need to be considered. Epigenetic modifications affect gene expression without changing the deoxyribonucleic acid sequence. In addition, environmental exposure during critical periods of development can affect the epigenetic tags and lead to obesity. A deeper understanding of the epigenetic mechanisms underlying obesity can aid in prevention based on lifestyle changes. This review focuses on the role of epigenetic modifications in the development of obesity and related conditions.
Collapse
Affiliation(s)
- O. M. Drapkina
- National Research Center for Therapy and Preventive Medicine
| | - O. T. Kim
- National Research Center for Therapy and Preventive Medicine
| |
Collapse
|
22
|
Khoshnejat M, Kavousi K, Banaei-Moghaddam AM, Moosavi-Movahedi AA. Unraveling the molecular heterogeneity in type 2 diabetes: a potential subtype discovery followed by metabolic modeling. BMC Med Genomics 2020; 13:119. [PMID: 32831068 PMCID: PMC7444195 DOI: 10.1186/s12920-020-00767-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 08/12/2020] [Indexed: 11/22/2022] Open
Abstract
Background Type 2 diabetes mellitus (T2DM) is a complex multifactorial disease with a high prevalence worldwide. Insulin resistance and impaired insulin secretion are the two major abnormalities in the pathogenesis of T2DM. Skeletal muscle is responsible for over 75% of the glucose uptake and plays a critical role in T2DM. Here, we sought to provide a better understanding of the abnormalities in this tissue. Methods The muscle gene expression patterns were explored in healthy and newly diagnosed T2DM individuals using supervised and unsupervised classification approaches. Moreover, the potential of subtyping T2DM patients was evaluated based on the gene expression patterns. Results A machine-learning technique was applied to identify a set of genes whose expression patterns could discriminate diabetic subjects from healthy ones. A gene set comprising of 26 genes was found that was able to distinguish healthy from diabetic individuals with 94% accuracy. In addition, three distinct clusters of diabetic patients with different dysregulated genes and metabolic pathways were identified. Conclusions This study indicates that T2DM is triggered by different cellular/molecular mechanisms, and it can be categorized into different subtypes. Subtyping of T2DM patients in combination with their real clinical profiles will provide a better understanding of the abnormalities in each group and more effective therapeutic approaches in the future.
Collapse
Affiliation(s)
- Maryam Khoshnejat
- Laboratory of Complex Biological Systems and Bioinformatics (CBB), Department of Bioinformatics, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran.,The UNESCO Chair on Interdisciplinary Research in Diabetes, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Kaveh Kavousi
- Laboratory of Complex Biological Systems and Bioinformatics (CBB), Department of Bioinformatics, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran. .,The UNESCO Chair on Interdisciplinary Research in Diabetes, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran.
| | - Ali Mohammad Banaei-Moghaddam
- The UNESCO Chair on Interdisciplinary Research in Diabetes, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran.,Laboratory of Genomics and Epigenomics (LGE), Department of Biochemistry, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Ali Akbar Moosavi-Movahedi
- The UNESCO Chair on Interdisciplinary Research in Diabetes, Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran.,Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| |
Collapse
|
23
|
Doaei S, Kalantari N, Izadi P, Salonurmi T, Jarrahi AM, Rafieifar S, Azizi Tabesh G, Rahimzadeh G, Gholamalizadeh M, Goodarzi MO. Interactions between macro-nutrients' intake, FTO and IRX3 gene expression, and FTO genotype in obese and overweight male adolescents. Adipocyte 2019; 8:386-391. [PMID: 31771407 PMCID: PMC6948981 DOI: 10.1080/21623945.2019.1693745] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 10/10/2019] [Accepted: 11/08/2019] [Indexed: 10/25/2022] Open
Abstract
This study is the first to identify the effects of FTO genotype on the interactions between the level of macro-nutrients intake and the expression level of fat mass and obesity associated (FTO) and homeobox transcription factor iriquois-3 (IRX3) genes This longitudinal study was carried out on 84 overweight and obese adolescent boys in Tehran, Iran. The rs9930506 SNP in FTO was genotyped at baseline and the level of FTO and IRX3 expression in PBMCs and macro-nutrients' intake were assessed at baseline and after 18 weeks of the intervention. The results identified that the higher carbohydrates intake significantly up-regulated the FTO gene (P = 0.001) and down-regulated the IRX3 gene (P = 0.01). Protein intake up-regulated the FTO gene (P = 0.001). In carriers of GG genotype of FTO gene, the amount of dietary carbohydrate had a positive association with FTO gene expression (p = 0.001, and p = 0.04, respectively). In AA/AG carriers, dietary protein was positively associated with FTO gene expression (p = 0.001) and dietary carbohydrate was negatively associated with IRX3 gene expression (P = 0.04). Therefore, dietary carbohydrateseems to be associated with FTO and IRX3 genes expression. These associations are influenced by FTO genotype.
Collapse
Affiliation(s)
- Saeid Doaei
- Research Center of Health and Environment, Guilan University of Medical Sciences, Rasht, Iran
- Student Research Committee, Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Naser Kalantari
- Department of Community Nutrition, School of Nutrition and Food Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Pantea Izadi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Tuire Salonurmi
- Department of Internal Medicine, Oulu University Hospital and University of Oulu, Oulu, Finland
| | | | - Shahram Rafieifar
- Health Promotion and Education Department, Ministry of Health, Tehran, Iran
| | - Ghasem Azizi Tabesh
- Department of Medical Genetics, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghazaleh Rahimzadeh
- Institute for Intelligent Systems Research and Innovation (IISRI), Deakin University, Geelong Waurn Ponds, Australia
| | - Maryam Gholamalizadeh
- Student Research Committee, Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mark O. Goodarzi
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
24
|
Agurs-Collins T, Ross SA, Dunn BK. The Many Faces of Obesity and Its Influence on Breast Cancer Risk. Front Oncol 2019; 9:765. [PMID: 31555578 PMCID: PMC6737012 DOI: 10.3389/fonc.2019.00765] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/29/2019] [Indexed: 12/12/2022] Open
Abstract
Obesity is associated with increased risk of breast and other cancers. However, the complexity of the underlying mechanisms, together with the interplay of diet and physical activity—contributing to energy balance—and the role of adipose tissue, pose challenges to our understanding of the basis of this increased risk. Epidemiologic studies have documented a higher obesity prevalence in US black women compared to white women. Elucidation of the contribution of potential biological differences among racially distinct groups to their differences in breast cancer (BC) risk and mortality have been topics of considerable interest in recent years. The racial and ethnic variation in body fat distribution may account for at least part of the differences in breast cancer rates in these populations. Yet, while black women exhibit higher rates of obesity compared to white women, this does not translate directly into higher rates of BC. In fact, overall, BC in black women occurs with a lower incidence than BC in white women. Obesity is a known risk factor for postmenopausal breast cancer, and growing evidence suggests that abdominal obesity, also known as central obesity, may increase risk for triple negative breast cancer, which is more common in premenopausal women. The positive association of postmenopausal BC risk and specifically estrogen receptor (ER)-positive BC, is presumably due largely to accumulation of estrogen in the adipose tissue of the breast and other tissues. Of the two main types of adipose tissue—subcutaneous and visceral—visceral adipocytes are more active metabolically. Such adipose tissue harbors multiple molecular entities that promote carcinogenesis: endocrine molecules/hormones, immunologic factors, inflammatory cytokines, metabolic alterations, and other components of the microenvironment. Expression of these culpable entities is largely regulated by epigenetic mechanisms. The interrelationship between these entities and drivers of epigenetic alteration are critical to the regulation of pathways connecting obesity and cancer risk. Initiatives to counteract the carcinogenic effects of obesity have primarily involved modulation of energy balance by diet. However, targeting of specific molecular abnormalities characterizing adiposity offers an alternative approach to preventing cancer. Our goal in this review is to first discuss the major mechanisms contributing to the obesity-breast cancer link. We will also consider race, specifically black/white differences, as they relate to the association of obesity with breast cancer risk. Then we will enumerate strategies targeting these mechanisms to reduce BC risk, in large part by way of dietary interventions with potential to mitigate the cancer-promoting components of adiposity.
Collapse
Affiliation(s)
- Tanya Agurs-Collins
- Division of Cancer Control and Population Sciences, National Cancer Institute, Rockville, MD, United States
| | - Sharon A Ross
- Division of Cancer Prevention, National Cancer Institute, Rockville, MD, United States
| | - Barbara K Dunn
- Division of Cancer Prevention, National Cancer Institute, Rockville, MD, United States
| |
Collapse
|
25
|
Mao Z, Yang Q, Yin W, Su W, Lin H, Feng M, Pan K, Yin Y, Zhang W. ETV5 regulates GOAT/ghrelin system in an mTORC1-dependent manner. Mol Cell Endocrinol 2019; 485:72-80. [PMID: 30735697 DOI: 10.1016/j.mce.2019.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 02/01/2019] [Accepted: 02/04/2019] [Indexed: 12/11/2022]
Abstract
Ghrelin, a 28 amino acid peptide hormone, regulates multiple important metabolic functions. Its acylation by ghrelin-O-acyl-transferase enzyme (GOAT) is required for binding to and activating its receptor, the growth hormone secretagogue receptor 1a. Mechanism underlying the regulation of GOAT and acyl ghrelin remains unclear. The present study demonstrated that ETV5 could transactivate GOAT promoter region and increase its expression, leading to subsequent increase in the production of acyl ghrelin. mTORC1 modulated ETV5 expression levels, likely via altering its protein stability, in the murine hypothalamic CLU122 cells and in mice. Moreover, ETV5 mediated the effects of mTORC1 signaling on the expression level of acyl ghrelin. Our study suggests a novel mTORC1-ETV5-GOAT/ghrelin axis in the regulation of ghrelin system. ETV5 may be a key regulator of mTORC1-GOAT/ghrelin axis in ghrelin producing cells and a potential therapeutic target for organism energy imbalance.
Collapse
Affiliation(s)
- Zhuo Mao
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong province, 518000, China.
| | - Qing Yang
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong province, 518000, China
| | - Wenzhen Yin
- Department of Physiology and Pathophysiology, School of Basic Science, Peking University Health Science Center, Beijing, 100191, China
| | - Wen Su
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong province, 518000, China
| | - Hui Lin
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong province, 518000, China
| | - Mingji Feng
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong province, 518000, China
| | - Ke Pan
- Institute for Advanced Study, Shenzhen University, Shenzhen, Guangdong province, 518000, China
| | - Yue Yin
- Department of Physiology and Pathophysiology, School of Basic Science, Peking University Health Science Center, Beijing, 100191, China
| | - Weizhen Zhang
- Center for Diabetes, Obesity and Metabolism, Department of Physiology, Shenzhen University Health Science Center, Shenzhen, Guangdong province, 518000, China; Department of Physiology and Pathophysiology, School of Basic Science, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
26
|
García-Carrizo F, Picó C, Rodríguez AM, Palou A. High-Esterified Pectin Reverses Metabolic Malprogramming, Improving Sensitivity to Adipostatic/Adipokine Hormones. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:3633-3642. [PMID: 30855142 DOI: 10.1021/acs.jafc.9b00296] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Detrimental metabolic programming has become a determinant factor in obesity propensity and the development of metabolic disorders; therefore, the search of nutritional strategies to reverse it is very relevant. Pectin is a prebiotic with health-promoting effects, such as control of glucose homeostasis and lipid metabolism, although other possible health effects and the prevention of obesity have been poorly studied. We studied the effects of chronic physiological supplementation with high-esterified pectin (HEP) in the reversion of metabolic nutrition-sensitive malprogramming associated with gestational undernutrition. As a model of nutrition-sensitive malprogramming, we used the progeny of rats with mild calorie restriction (CR) during pregnancy and analyzed their performance under metabolic stress (high-sucrose diet). We focused on the study of the sensitivity to the main adipostatic/adipokine hormones, i.e., leptin, insulin, and adiponectin, at both peripheral (liver and circulating parameters) and central (hypothalamus) levels. Our main findings suggest that chronic HEP supplementation is able to prevent weight/fat gain, to substantially reverse the detrimental malprogramming caused by the CR condition, to improve general health circulating markers, to modulate oxidative/lipogenic balance in the liver and energy metabolism regulators in the hypothalamus, and to restore/improve adipostatic/adipokine sensitivity affected by maternal calorie restriction, both peripherally and centrally. HEP stands out as a food component potentially useful against the development of metabolic disorders and obesity.
Collapse
Affiliation(s)
- Francisco García-Carrizo
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics) , University of the Balearic Islands (UIB) and Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBEROBN) , 07122 Palma de Mallorca , Spain
| | - Catalina Picó
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics) , University of the Balearic Islands (UIB) and Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBEROBN) , 07122 Palma de Mallorca , Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa) , 07120 Palma de Mallorca , Spain
| | - Ana María Rodríguez
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics) , University of the Balearic Islands (UIB) and Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBEROBN) , 07122 Palma de Mallorca , Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa) , 07120 Palma de Mallorca , Spain
| | - Andreu Palou
- Laboratory of Molecular Biology, Nutrition and Biotechnology (Nutrigenomics) , University of the Balearic Islands (UIB) and Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBEROBN) , 07122 Palma de Mallorca , Spain
- Institut d'Investigació Sanitària Illes Balears (IdISBa) , 07120 Palma de Mallorca , Spain
| |
Collapse
|
27
|
Liu L, Yi J, Ray WK, Vu LT, Helm RF, Siegel PB, Cline MA, Gilbert ER. Fasting differentially alters the hypothalamic proteome of chickens from lines with the propensity to be anorexic or obese. Nutr Diabetes 2019; 9:13. [PMID: 30931934 PMCID: PMC6443654 DOI: 10.1038/s41387-019-0081-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Revised: 02/27/2019] [Accepted: 03/12/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The hypothalamus is the ultimate modulator of appetite and energy balance and therefore sensitive to changes in nutritional state. Chicks from lines selected for low (LWS) and high (HWS) body weight are hypophagic and compulsive eaters, respectively, and differ in their propensity to become obese and in their hypothalamic mRNA response to fasting. METHODS As fasting-induced changes in hypothalamic proteins are unknown, we investigated the hypothalamic proteomes of 5-day old LWS and HWS chicks in the fed and fasted states using a label-free liquid chromatography-tandem mass spectrometry (LC-MS/MS) approach. RESULTS A total of 744 proteins were identified in the chicken hypothalamus, and 268 differentially abundant proteins were identified among four pairwise comparisons. Ninety-five proteins were associated with the response to fasting in HWS chicks, and 23 proteins were associated with the response to fasting in LWS chicks. Fasting-responsive proteins in HWS chicks were significantly enriched in ATP metabolic processes, glyoxylate/dicarboxylate metabolism, and ribosome function. There was no enrichment for any pathways in LWS chicks in response to fasting. In the fasted and fed states, 159 and 119 proteins differed between HWS and LWS, respectively. Oxidative phosphorylation, citric acid cycle, and carbon metabolism were the main pathways associated with differences between the two lines of chicks. Enzymes associated with metabolic pathways differed between HWS and LWS in both nutritional states, including fumarase, aspartate aminotransferase, mitochondrial GOT2, 3-hydroxyisobutyrate dehydrogenase, chondrogenesis associated lipocalin, sialic acid synthase, arylamine N-acetyltransferase, pineal gland isozyme NAT-3, and succinate dehydrogenase [ubiquinone] flavoprotein subunit, mitochondrial. CONCLUSIONS These results provide insights into the hypothalamic metabolic pathways that are affected by nutritional status and the regulation of appetite and eating behavior.
Collapse
Affiliation(s)
- Lingbin Liu
- College of Animal Science and Technology, Southwest University, Chongqing, P.R. China
| | - Jiaqing Yi
- Virginia Tech, Department of Animal and Poultry Sciences, Blacksburg, VA, USA
| | - W Keith Ray
- Virginia Tech, Department of Biochemistry, Blacksburg, VA, USA
| | - Lucas T Vu
- Virginia Tech, Department of Chemical Engineering, Blacksburg, VA, USA
| | - Richard F Helm
- Virginia Tech, Department of Biochemistry, Blacksburg, VA, USA
| | - Paul B Siegel
- Virginia Tech, Department of Animal and Poultry Sciences, Blacksburg, VA, USA
| | - Mark A Cline
- Virginia Tech, Department of Animal and Poultry Sciences, Blacksburg, VA, USA
| | - Elizabeth R Gilbert
- Virginia Tech, Department of Animal and Poultry Sciences, Blacksburg, VA, USA.
| |
Collapse
|
28
|
Rohde K, Keller M, la Cour Poulsen L, Blüher M, Kovacs P, Böttcher Y. Genetics and epigenetics in obesity. Metabolism 2019; 92:37-50. [PMID: 30399374 DOI: 10.1016/j.metabol.2018.10.007] [Citation(s) in RCA: 225] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 10/15/2018] [Accepted: 10/21/2018] [Indexed: 12/20/2022]
Abstract
Obesity is among the most threatening health burdens worldwide and its prevalence has markedly increased over the last decades. Obesity maybe considered a heritable trait. Identifications of rare cases of monogenic obesity unveiled that hypothalamic circuits and the brain-adipose axis play an important role in the regulation of energy homeostasis, appetite, hunger and satiety. For example, mutations in the leptin gene cause obesity through almost unsuppressed overeating. Common (multifactorial) obesity, most likely resulting from a concerted interplay of genetic, epigenetic and environmental factors, is clearly linked to genetic predisposition by multiple risk variants, which, however only account for a minor part of the general BMI variability. Although GWAS opened new avenues in elucidating the complex genetics behind common obesity, understanding the biological mechanisms relative to the specific risk contributing to obesity remains poorly understood. Non-genetic factors such as eating behavior or physical activity strongly modulate the individual risk for developing obesity. These factors may interact with genetic predisposition for obesity through epigenetic mechanisms. Thus, here, we review the current knowledge about monogenic and common (multifactorial) obesity highlighting the important recent advances in our knowledge on how epigenetic regulation is involved in the etiology of obesity.
Collapse
Affiliation(s)
- Kerstin Rohde
- Leipzig University Medical Center, IFB Adiposity Diseases, Leipzig 04103, Germany; University of Oslo, Institute of Clinical Medicine, Oslo 0316, Norway.
| | - Maria Keller
- Leipzig University Medical Center, IFB Adiposity Diseases, Leipzig 04103, Germany.
| | - Lars la Cour Poulsen
- Akershus University Hospital, Department of Clinical Molecular Biology, Medical Division, Lørenskog 1478, Norway.
| | - Matthias Blüher
- Department of Medicine, University of Leipzig, Leipzig 04103, Germany.
| | - Peter Kovacs
- Leipzig University Medical Center, IFB Adiposity Diseases, Leipzig 04103, Germany.
| | - Yvonne Böttcher
- Leipzig University Medical Center, IFB Adiposity Diseases, Leipzig 04103, Germany; University of Oslo, Institute of Clinical Medicine, Oslo 0316, Norway; Akershus University Hospital, Department of Clinical Molecular Biology, Medical Division, Lørenskog 1478, Norway.
| |
Collapse
|
29
|
Depression and obesity: evidence of shared biological mechanisms. Mol Psychiatry 2019; 24:18-33. [PMID: 29453413 DOI: 10.1038/s41380-018-0017-5] [Citation(s) in RCA: 626] [Impact Index Per Article: 104.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 11/13/2017] [Accepted: 12/06/2017] [Indexed: 12/17/2022]
Abstract
Depression and obesity are common conditions with major public health implications that tend to co-occur within individuals. The relationship between these conditions is bidirectional: the presence of one increases the risk for developing the other. It has thus become crucial to gain a better understanding of the mechanisms responsible for the intertwined downward physiological spirals associated with both conditions. The present review focuses specifically on shared biological pathways that may mechanistically explain the depression-obesity link, including genetics, alterations in systems involved in homeostatic adjustments (HPA axis, immuno-inflammatory activation, neuroendocrine regulators of energy metabolism including leptin and insulin, and microbiome) and brain circuitries integrating homeostatic and mood regulatory responses. Furthermore, the review addresses interventional opportunities and questions to be answered by future research that will enable a comprehensive characterization and targeting of the biological links between depression and obesity.
Collapse
|
30
|
Kadumuri RV, Janga SC. Epitranscriptomic Code and Its Alterations in Human Disease. Trends Mol Med 2018; 24:886-903. [PMID: 30120023 DOI: 10.1016/j.molmed.2018.07.010] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/23/2018] [Accepted: 07/24/2018] [Indexed: 02/07/2023]
Abstract
Innovations in epitranscriptomics have resulted in the identification of more than 160 RNA modifications to date. These developments, together with the recent discovery of writers, readers, and erasers of modifications occurring across a wide range of RNAs and tissue types, have led to a surge in integrative approaches for transcriptome-wide mapping of modifications and protein-RNA interaction profiles of epitranscriptome players. RNA modification maps and crosstalk between them have begun to elucidate the role of modifications as signaling switches, entertaining the notion of an epitranscriptomic code as a driver of the post-transcriptional fate of RNA. Emerging single-molecule sequencing technologies and development of antibodies specific to various RNA modifications could enable charting of transcript-specific epitranscriptomic marks across cell types and their alterations in disease.
Collapse
Affiliation(s)
- Rajashekar Varma Kadumuri
- Department of BioHealth Informatics, School of Informatics and Computing, Walker Plaza Building, Indiana University-Purdue University Indianapolis, 719 Indiana Avenue, Suite 319, Indianapolis, IN 46202, USA
| | - Sarath Chandra Janga
- Department of BioHealth Informatics, School of Informatics and Computing, Walker Plaza Building, Indiana University-Purdue University Indianapolis, 719 Indiana Avenue, Suite 319, Indianapolis, IN 46202, USA; Department of Medical and Molecular Genetics, Medical Research and Library Building, Indiana University School of Medicine, 975 West Walnut Street, Indianapolis, IN 46202, USA; Centre for Computational Biology and Bioinformatics, 5021 Health Information and Translational Sciences, Indiana University School of Medicine, 410 West 10th Street, Indianapolis, IN 46202, USA.
| |
Collapse
|
31
|
Engel M, Chen A. The emerging role of mRNA methylation in normal and pathological behavior. GENES BRAIN AND BEHAVIOR 2017; 17:e12428. [PMID: 29027751 DOI: 10.1111/gbb.12428] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 10/02/2017] [Accepted: 10/09/2017] [Indexed: 12/11/2022]
Abstract
Covalent RNA modifications were recently rediscovered as abundant RNA chemical tags. Similarly to DNA epigenetic modifications, they have been proposed as essential regulators of gene expression. Here we focus on 3 of the most abundant adenosine methylations: N6-methyladenosine (m6 A), N6,2'-O-dimethyladenosine (m6 Am) and N1-methyladenosine (m1 A). We review the potential role of these modifications on mature mRNA in regulating gene expression within the adult brain, nervous system function and normal and pathological behavior. Dynamic mRNA modifications, summarized as the epitranscriptome, regulate transcript maturation, translation and decay, and thus crucially determine gene expression beyond primary transcription regulation. However, the extent of this regulation in the healthy and maladapted adult brain is poorly understood. Analyzing this novel layer of gene expression control in addition to epigenetics and posttranslational regulation of proteins will be highly relevant for understanding the molecular underpinnings of behavior and psychiatric disorders.
Collapse
Affiliation(s)
- M Engel
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - A Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany.,Department of Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
32
|
Genome-wide association study for feed efficiency and growth traits in U.S. beef cattle. BMC Genomics 2017; 18:386. [PMID: 28521758 PMCID: PMC5437562 DOI: 10.1186/s12864-017-3754-y] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 05/03/2017] [Indexed: 11/13/2022] Open
Abstract
Background Single nucleotide polymorphism (SNP) arrays for domestic cattle have catalyzed the identification of genetic markers associated with complex traits for inclusion in modern breeding and selection programs. Using actual and imputed Illumina 778K genotypes for 3887 U.S. beef cattle from 3 populations (Angus, Hereford, SimAngus), we performed genome-wide association analyses for feed efficiency and growth traits including average daily gain (ADG), dry matter intake (DMI), mid-test metabolic weight (MMWT), and residual feed intake (RFI), with marker-based heritability estimates produced for all traits and populations. Results Moderate and/or large-effect QTL were detected for all traits in all populations, as jointly defined by the estimated proportion of variance explained (PVE) by marker effects (PVE ≥ 1.0%) and a nominal P-value threshold (P ≤ 5e-05). Lead SNPs with PVE ≥ 2.0% were considered putative evidence of large-effect QTL (n = 52), whereas those with PVE ≥ 1.0% but < 2.0% were considered putative evidence for moderate-effect QTL (n = 35). Identical or proximal lead SNPs associated with ADG, DMI, MMWT, and RFI collectively supported the potential for either pleiotropic QTL, or independent but proximal causal mutations for multiple traits within and between the analyzed populations. Marker-based heritability estimates for all investigated traits ranged from 0.18 to 0.60 using 778K genotypes, or from 0.17 to 0.57 using 50K genotypes (reduced from Illumina 778K HD to Illumina Bovine SNP50). An investigation to determine if QTL detected by 778K analysis could also be detected using 50K genotypes produced variable results, suggesting that 50K analyses were generally insufficient for QTL detection in these populations, and that relevant breeding or selection programs should be based on higher density analyses (imputed or directly ascertained). Conclusions Fourteen moderate to large-effect QTL regions which ranged from being physically proximal (lead SNPs ≤ 3Mb) to fully overlapping for RFI, DMI, ADG, and MMWT were detected within and between populations, and included evidence for pleiotropy, proximal but independent causal mutations, and multi-breed QTL. Bovine positional candidate genes for these traits were functionally conserved across vertebrate species. Electronic supplementary material The online version of this article (doi:10.1186/s12864-017-3754-y) contains supplementary material, which is available to authorized users.
Collapse
|
33
|
Mechaly AS, Richardson E, Rinkwitz S. Activity of etv5a and etv5b genes in the hypothalamus of fasted zebrafish is influenced by serotonin. Gen Comp Endocrinol 2017; 246:233-240. [PMID: 28041791 DOI: 10.1016/j.ygcen.2016.12.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/03/2016] [Accepted: 12/19/2016] [Indexed: 11/28/2022]
Abstract
Serotonin has been implicated in the inhibition of food intake in vertebrates. However, the mechanisms through which serotonin acts has yet to be elucidated. Recently, ETV5 (ets variant gene 5) has been associated with obesity and food intake control mechanisms in mammals. We have analyzed a putative physiological function of the two etv5 paralogous genes (etv5a and etv5b) in neuronal food intake control in adult zebrafish that have been exposed to different nutritional conditions. A feeding assay was established and fluoxetine, a selective serotonin re-uptake inhibitor (SSRI), was applied. Gene expression changes in the hypothalamus were determined using real-time PCR. Fasting induced an up-regulation of etv5a and etv5b in the hypothalamus, whereas increased serotonin levels in the fasted fish counteracted the increase in expression. To investigate potential mechanisms the expression of further food intake control genes was determined. The results show that an increase of serotonin in fasting fish causes a reduction in the activity of genes stimulating food intake. This is in line with a previously demonstrated anorexigenic function of serotonin. Our results suggest that obesity-associated ETV5 has a food intake stimulating function and that this function is modulated through serotonin.
Collapse
Affiliation(s)
- Alejandro S Mechaly
- Dept. of Physiology, Sydney Medical School, University of Sydney, Camperdown 2050, Australia.
| | - Ebony Richardson
- Dept. of Physiology, Sydney Medical School, University of Sydney, Camperdown 2050, Australia
| | - Silke Rinkwitz
- Dept. of Physiology, Sydney Medical School, University of Sydney, Camperdown 2050, Australia.
| |
Collapse
|
34
|
Doaei S, Kalantari N, Mohammadi NK, Tabesh GA, Gholamalizadeh M. Macronutrients and the FTO gene expression in hypothalamus; a systematic review of experimental studies. Indian Heart J 2017; 69:277-281. [PMID: 28460778 PMCID: PMC5414942 DOI: 10.1016/j.ihj.2017.01.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2016] [Revised: 12/28/2016] [Accepted: 01/17/2017] [Indexed: 12/31/2022] Open
Abstract
The various studies have examined the relationship between FTO gene expression and macronutrients levels. In order to obtain better viewpoint from this interactions, all of existing studies were reviewed systematically. All published papers have been obtained and reviewed using standard and sensitive keywords from databases such as CINAHL, Embase, PubMed, PsycInfo, and the Cochrane, from 1990 to 2016. The results indicated that all of 6 studies that met the inclusion criteria (from a total of 428 published article) found FTO gene expression changes at short-term follow-ups. Four of six studies found an increased FTO gene expression after calorie restriction, while two of them indicated decreased FTO gene expression. The effect of protein, carbohydrate and fat were separately assessed and suggested by all of six studies. In Conclusion, The level of FTO gene expression in hypothalamus is related to macronutrients levels. Future research should evaluate the long-term impact of dietary interventions.
Collapse
Affiliation(s)
- Saeid Doaei
- Student's Research Committee, National Nutrition and Food Technology Research Institute, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Naser Kalantari
- Department of Community Nutrition, Faculty of Nutrition Sciences and Food Technology, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Ghasem Azizi Tabesh
- Department of Human Genetics, Faculty of Human Genetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Gholamalizadeh
- Student's Research Committee, Cancer Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
35
|
She K, Huang J, Zhou H, Huang T, Chen G, He J. lncRNA-SNHG7 promotes the proliferation, migration and invasion and inhibits apoptosis of lung cancer cells by enhancing the FAIM2 expression. Oncol Rep 2016; 36:2673-2680. [PMID: 27666964 DOI: 10.3892/or.2016.5105] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 07/11/2016] [Indexed: 11/06/2022] Open
Abstract
There is growing evidence that long non-coding RNAs (lncRNAs) are related to cancer development. In the present study, we found that the expression levels of lncRNA-SNHG7 mRNA and protein obviously increased in lung cancer tissues compared to adjacent non-cancerous tissues. Simultaneously, the expression levels of Fas apoptotic inhibitory molecule 2 (FAIM2) also increased in lung cancer tissues. In addition, lncRNA-SNHG7 was of positive relevance with FAIM2 in human lung cancer tissues. Silence of lncRNA‑SNHG7 by siRNA repressed the level of FAIM2 protein and suppressed cell proliferation, migration and invasion and accelerated apoptosis of A594 cells in vitro. Furthermore, silence of FAIM2 by siRNA generated a phenotype similar to silence of lncRNA-SNHG7 by siRNA. Therefore, our research showed that lncRNA-SNHG7 promotes the proliferation, migration and invasion, and inhibits apoptosis of lung cancer cells by enhancing the FAIM2 expression, suggesting that lncRNA-SNHG7 as a key regulator of gene expression, may be a promising therapeutic strategy for the treatment of lung cancer. It may improve the understanding of their biogenesis and function of lung cancer and further provide the theoretical fundamental basis for cancer pathogenesis and treatment.
Collapse
Affiliation(s)
- Kelin She
- Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Jun Huang
- State Key Laboratory of Respiratory Disease, Guangzhou, Guangdong, P.R. China
| | - Huaping Zhou
- Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Tonghai Huang
- Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Guojun Chen
- Southern Medical University, Guangzhou, Guangdong, P.R. China
| | - Jianxing He
- Southern Medical University, Guangzhou, Guangdong, P.R. China
| |
Collapse
|
36
|
Xiang L, Wu H, Pan A, Patel B, Xiang G, Qi L, Kaplan RC, Hu F, Wylie-Rosett J, Qi Q. FTO genotype and weight loss in diet and lifestyle interventions: a systematic review and meta-analysis. Am J Clin Nutr 2016; 103:1162-70. [PMID: 26888713 PMCID: PMC4807705 DOI: 10.3945/ajcn.115.123448] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 01/20/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Studies have suggested that the fat mass and obesity-associated (FTO) genotype is associated with individual variability in weight loss in response to diet/lifestyle interventions, but results are inconsistent. OBJECTIVE We aimed to provide a summary of the literature evaluating the relation between the FTO genotype and weight loss in response to diet/lifestyle interventions. DESIGN A search of English-language articles in the PubMed and Embase databases (through 30 April 2015) was performed. Eligible studies were diet/lifestyle weight-loss intervention studies conducted in adults that reported changes in body weight or body mass index (BMI) by the FTO variant rs9939609 (or its proxy). Differences in weight loss between FTO genotypes across studies were pooled with the use of fixed-effect models. RESULTS A meta-analysis of 10 studies (comprising 6951 participants) that reported the results of additive genetic models showed that individuals with the FTO TA genotype and AA genotype (those with the obesity-predisposing A allele) had 0.18-kg (95% CI: -0.09-, 0.45-kg;P= 0.19; NS) and 0.44-kg (95% CI: 0.09-, 0.79-kg;P= 0.015) greater weight loss, respectively, than those with the TT genotype. A meta-analysis of 14 studies (comprising 7700 participants) that reported the results of dominant genetic models indicated a 0.20-kg (-0.43-, 0.04-kg) greater weight loss in the TA/AA genotype than in the TT genotype (P= 0.10). In addition, differences in weight loss between the AA genotype and TT genotype were significant in studies with a diet intervention only, adjustment for baseline BMI or body weight, and several other subgroups. However, the relatively small number of studies limited these stratified analyses, and there was no statistically significant difference between subgroups. CONCLUSIONS This meta-analysis suggests that individuals carrying the homozygous FTO obesity-predisposing allele may lose more weight through diet/lifestyle interventions than noncarriers. Our data provide evidence for genetic variability in response to diet/lifestyle interventions on weight loss, although clinical applications of these findings need further investigations.
Collapse
Affiliation(s)
- Lingwei Xiang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Hongyu Wu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - An Pan
- School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bhakti Patel
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Guangda Xiang
- Department of Endocrinology, Wuhan General Hospital of Guangzhou Command, Wuhan, China; and
| | - Lu Qi
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Robert C Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Frank Hu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Judith Wylie-Rosett
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY;
| |
Collapse
|
37
|
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: 1.1] [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
|
38
|
Qi Q, Downer MK, Kilpeläinen TO, Taal HR, Barton SJ, Ntalla I, Standl M, Boraska V, Huikari V, Kiefte-de Jong JC, Körner A, Lakka TA, Liu G, Magnusson J, Okuda M, Raitakari O, Richmond R, Scott RA, Bailey MES, Scheuermann K, Holloway JW, Inskip H, Isasi CR, Mossavar-Rahmani Y, Jaddoe VWV, Laitinen J, Lindi V, Melén E, Pitsiladis Y, Pitkänen N, Snieder H, Heinrich J, Timpson NJ, Wang T, Yuji H, Zeggini E, Dedoussis GV, Kaplan RC, Wylie-Rosett J, Loos RJF, Hu FB, Qi L. Dietary Intake, FTO Genetic Variants, and Adiposity: A Combined Analysis of Over 16,000 Children and Adolescents. Diabetes 2015; 64:2467-76. [PMID: 25720386 PMCID: PMC4876751 DOI: 10.2337/db14-1629] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2014] [Accepted: 02/12/2015] [Indexed: 12/26/2022]
Abstract
The FTO gene harbors variation with the strongest effect on adiposity and obesity risk. Previous data support a role for FTO variation in influencing food intake. We conducted a combined analysis of 16,094 boys and girls aged 1-18 years from 14 studies to examine the following: 1) the association between the FTO rs9939609 variant (or a proxy) and total energy and macronutrient intake; and 2) the interaction between the FTO variant and dietary intake, and the effect on BMI. We found that the BMI-increasing allele (minor allele) of the FTO variant was associated with increased total energy intake (effect per allele = 14.3 kcal/day [95% CI 5.9, 22.7 kcal/day], P = 6.5 × 10(-4)), but not with protein, carbohydrate, or fat intake. We also found that protein intake modified the association between the FTO variant and BMI (interactive effect per allele = 0.08 SD [0.03, 0.12 SD], P for interaction = 7.2 × 10(-4)): the association between FTO genotype and BMI was much stronger in individuals with high protein intake (effect per allele = 0.10 SD [0.07, 0.13 SD], P = 8.2 × 10(-10)) than in those with low intake (effect per allele = 0.04 SD [0.01, 0.07 SD], P = 0.02). Our results suggest that the FTO variant that confers a predisposition to higher BMI is associated with higher total energy intake, and that lower dietary protein intake attenuates the association between FTO genotype and adiposity in children and adolescents.
Collapse
Affiliation(s)
- Qibin Qi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY Department of Nutrition, Harvard School of Public Health, Boston, MA
| | - Mary K Downer
- Department of Nutrition, Harvard School of Public Health, Boston, MA
| | - Tuomas O Kilpeläinen
- MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke's Hospital and University of Cambridge, Cambridge, U.K. The Novo Nordisk Foundation Center for Basic Metabolic Research, Section of Metabolic Genetics, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - H Rob Taal
- The Generation R Study Group, Erasmus Medical Center, Rotterdam, the Netherlands Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands Department of Pediatrics, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Sheila J Barton
- MRC Lifecourse Epidemiology Unit, Faculty of Medicine, University of Southampton, Southampton, U.K
| | - Ioanna Ntalla
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece Department of Health Sciences, University of Leicester, Leicester, U.K
| | - Marie Standl
- Institute of Epidemiology I, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Vesna Boraska
- Wellcome Trust Sanger Institute, Hixton, Cambridge, U.K. Department of Medical Biology, University of Split School of Medicine, Split, Croatia
| | - Ville Huikari
- Institute of Health Sciences, University of Oulu, Oulu, Finland
| | - Jessica C Kiefte-de Jong
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands Global Public Health, Leiden University College, Hague, the Netherlands
| | - Antje Körner
- Pediatric Research Center, Department of Women's & Child Health, University of Leipzig, Leipzig, Germany
| | - Timo A Lakka
- Institute of Biomedicine, Department of Physiology, University of Eastern Finland, Kuopio, Finland Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital and University of Eastern Finland, Kuopio, Finland Kuopio Research Institute of Exercise Medicine, Kuopio, Finland
| | - Gaifen Liu
- Department of Neurology, Beijing Tian Tan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Jessica Magnusson
- Institute of Environmental Medicine, Karolinska Institutet, and Sachs' Children and Youth Hospital, Stockholm, Sweden
| | - Masayuki Okuda
- Graduate School of Science and Engineering, Yamaguchi University, Ube, Japan
| | - Olli Raitakari
- The Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland Department of Clinical Physiology and Nuclear Medicine, Turku University Hospital, Turku, Finland
| | - Rebecca Richmond
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, U.K
| | - Robert A Scott
- MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke's Hospital and University of Cambridge, Cambridge, U.K
| | - Mark E S Bailey
- School of Life Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, U.K
| | - Kathrin Scheuermann
- Pediatric Research Center, Department of Women's & Child Health, University of Leipzig, Leipzig, Germany
| | - John W Holloway
- Human Genetics and Medical Genomics, Faculty of Medicine, University of Southampton, Southampton, U.K
| | - Hazel Inskip
- MRC Lifecourse Epidemiology Unit, Faculty of Medicine, University of Southampton, Southampton, U.K
| | - Carmen R Isasi
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Yasmin Mossavar-Rahmani
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Vincent W V Jaddoe
- The Generation R Study Group, Erasmus Medical Center, Rotterdam, the Netherlands Department of Epidemiology, Erasmus Medical Center, Rotterdam, the Netherlands Department of Pediatrics, Erasmus Medical Center, Rotterdam, the Netherlands
| | | | - Virpi Lindi
- Institute of Biomedicine, Department of Physiology, University of Eastern Finland, Kuopio, Finland
| | - Erik Melén
- Institute of Environmental Medicine, Karolinska Institutet, and Sachs' Children and Youth Hospital, Stockholm, Sweden
| | - Yannis Pitsiladis
- School of Life Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, U.K
| | - Niina Pitkänen
- The Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Harold Snieder
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands Georgia Prevention Center, Department of Pediatrics, Georgia Regents University, Augusta, GA
| | - Joachim Heinrich
- Institute of Epidemiology I, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | | | - Tao Wang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Hinoda Yuji
- Hokkaido Nursing College, Chuo-ku, Sapporo, Japan
| | | | - George V Dedoussis
- Department of Nutrition and Dietetics, School of Health Science and Education, Harokopio University, Athens, Greece
| | - Robert C Kaplan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Judith Wylie-Rosett
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Ruth J F Loos
- MRC Epidemiology Unit, Institute of Metabolic Science, Addenbrooke's Hospital and University of Cambridge, Cambridge, U.K. The Genetics of Obesity and Related Metabolic Traits Program, The Charles Bronfman Institute for Personalized Medicine, The Mindich Child Health and Development Institute, Department of Preventive Medicine, Icahn School of Medicine at Mount Sinai, New York City, NY
| | - Frank B Hu
- Department of Nutrition, Harvard School of Public Health, Boston, MA Department of Epidemiology, Harvard School of Public Health, Boston, MA Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| | - Lu Qi
- Department of Nutrition, Harvard School of Public Health, Boston, MA Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA
| |
Collapse
|
39
|
Wu L, Zhao X, Shen Y, Zhang MX, Yan Y, Hou D, Meng L, Liu J, Cheng H, Mi J. Promoter methylation of fas apoptotic inhibitory molecule 2 gene is associated with obesity and dyslipidaemia in Chinese children. Diab Vasc Dis Res 2015; 12:217-20. [PMID: 25696115 DOI: 10.1177/1479164114565630] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Fas apoptotic inhibitory molecule 2 (FAIM2) is an obesity-related gene, but the mechanisms by which FAIM2 is involved in obesity are not understood. Epigenetic alterations are important factors in the development of obesity. The purpose of this study was to investigate the potential associations of FAIM2 promoter methylation with obesity and components of dyslipidaemia in Chinese children. We studied FAIM2 promoter methylation in 59 obese and 39 lean children using the Sequenom MassARRAY platform. The methylation levels at 8 CpG sites in the FAIM2 promoter were significantly different between the obese and lean subjects, especially the methylation level at CpG site 500 (p = 0.01). The methylation levels at several of the examined CpG sites were significantly associated with dyslipidaemia and its components after adjusting for age, gender and body mass index (BMI). The methylation levels at two CpG sites (sites -362 and -360 and site -164) were highly significantly associated with high level of triglycerides (p = 0.00002 and 0.0009, respectively). This study provides the first evidence that the methylation levels of the FAIM2 promoter are significantly associated with obesity and are independently associated with dyslipidaemia and its components in Chinese children.
Collapse
Affiliation(s)
- Lijun Wu
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| | - Xiaoyuan Zhao
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| | - Yue Shen
- National Research Institute for Family Planning, Beijing, China
| | - Mei-Xian Zhang
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| | - Yinkun Yan
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| | - Dongqing Hou
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| | - Linghui Meng
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| | - Junting Liu
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| | - Hong Cheng
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| | - Jie Mi
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| |
Collapse
|
40
|
Wu L, Zhao X, Shen Y, Huang G, Zhang M, Yan Y, Hou D, Meng L, Liu J, Cheng H, Mi J. Influence of lifestyle on the FAIM2 promoter methylation between obese and lean children: a cohort study. BMJ Open 2015; 5:e007670. [PMID: 25922107 PMCID: PMC4420961 DOI: 10.1136/bmjopen-2015-007670] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE An obesity-related gene, Fas apoptotic inhibitory molecule 2 (FAIM2), is regulated by nutritional state and the methylation levels of the FAIM2 promoter are significantly associated with obesity. Lifestyle factors, such as sedentary behaviour and physical activity, might modify epigenetic patterns that have been related to obesity. Whether the molecular mechanisms by which FAIM2 affects obesity are involved in lifestyle is unclear. This study investigates the potential differences of the FAIM2 promoter methylation with sedentary behaviour and physical activity in obese and lean children. DESIGN Cohort study. SETTING Institute of Pediatrics in China. PARTICIPANTS 59 obese cases and 39 lean controls aged 8-18 years recruited from a cross-sectional survey of children from Beijing in 2013. PRIMARY AND SECONDARY OUTCOME MEASURES The FAIM2 promoter methylation was quantified using the Sequenom MassARRAY platform. Sedentary behaviour and physical activity were investigated using a questionnaire. The influences of different lifestyles on methylation variations in obese and lean children were examined by multiple linear regression. RESULTS The methylation levels at seven CpG sites of the FAIM2 promoter were significantly associated with sedentary behaviour, especially the methylation levels at site -975, site -413, sites -362 and -360, and sites -353 and -349 (p=0.00004, 0.00009, 0.0006 and 0.00005, respectively). There were significant differences between the methylation levels at four CpG sites in obese and lean participants with high or moderate physical activity level <150 min/week. CONCLUSIONS This study provides the first evidence that there are significant differences in the associations of the FAIM2 promoter methylation with sedentary behaviour and physical activity between obese and lean children. Our results suggest that lifestyle may possibly be mediating the process of the FAIM2 involved in obesity.
Collapse
Affiliation(s)
- Lijun Wu
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| | - Xiaoyuan Zhao
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| | - Yue Shen
- National Research Institute for Family Planning, Beijing, China
| | - Guimin Huang
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| | - Meixian Zhang
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| | - Yinkun Yan
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| | - Dongqing Hou
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| | - Linghui Meng
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| | - Junting Liu
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| | - Hong Cheng
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| | - Jie Mi
- Department of Epidemiology, Capital Institute of Pediatrics, Beijing, China
| |
Collapse
|
41
|
Gutierrez-Aguilar R, Thompson A, Marchand N, Dumont P, Woods SC, de Launoit Y, Seeley RJ, Ulrich-Lai YM. The obesity-associated transcription factor ETV5 modulates circulating glucocorticoids. Physiol Behav 2015; 150:38-42. [PMID: 25813907 DOI: 10.1016/j.physbeh.2015.03.027] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 03/03/2015] [Accepted: 03/20/2015] [Indexed: 12/16/2022]
Abstract
The transcription factor E-twenty-six version 5 (ETV5) has been linked with obesity in genome-wide association studies. Moreover, ETV5-deficient mice (knockout; KO) have reduced body weight, lower fat mass, and are resistant to diet-induced obesity, directly linking ETV5 to the regulation of energy balance and metabolism. ETV5 is expressed in hypothalamic brain regions that regulate both metabolism and HPA axis activity, suggesting that ETV5 may also modulate HPA axis function. In order to test this possibility, plasma corticosterone levels were measured in ETV5 KO and wildtype (WT) mice before (pre-stress) and after (post-stress) a mild stressor (intraperitoneal injection). ETV5 deficiency increased both pre- and post-stress plasma corticosterone, suggesting that loss of ETV5 elevated glucocorticoid tone. Consistent with this idea, ETV5 KO mice have reduced thymus weight, suggestive of increased glucocorticoid-induced thymic involution. ETV5 deficiency also decreased the mRNA expression of glucocorticoid receptor (GR), mineralocorticoid receptor (MR), and vasopressin receptor 1A in the hypothalamus, without altering vasopressin, corticotropin-releasing hormone, or oxytocin mRNA expression. In order to test whether reduced MR and GR expression affected glucocorticoid negative feedback, a dexamethasone suppression test was performed. Dexamethasone reduced plasma corticosterone in both ETV5 KO and WT mice, suggesting that glucocorticoid negative feedback was unaltered by ETV5 deficiency. In summary, these data suggest that the obesity-associated transcription factor ETV5 normally acts to diminish circulating glucocorticoids. This might occur directly via ETV5 actions on HPA-regulatory brain circuitry, and/or indirectly via ETV5-induced alterations in metabolic factors that then influence the HPA axis.
Collapse
Affiliation(s)
- Ruth Gutierrez-Aguilar
- Department of Internal Medicine, University of Cincinnati, United States; Laboratorio de Enfermedades Metabólicas Obesidad y Diabetes, Hospital Infantil de México Federico Gómez, Mexico.
| | - Abigail Thompson
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, 2170 East Galbraith Road, Cincinnati, OH 45237, United States
| | - Nathalie Marchand
- UMR8161 CNRS, Université de Lille, Institut Pasteur de Lille, Institut de Biologie de Lille, 1 Rue Calmette, 59021 Lille CEDEX, France
| | - Patrick Dumont
- UMR8161 CNRS, Université de Lille, Institut Pasteur de Lille, Institut de Biologie de Lille, 1 Rue Calmette, 59021 Lille CEDEX, France
| | - Stephen C Woods
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, 2170 East Galbraith Road, Cincinnati, OH 45237, United States
| | - Yvan de Launoit
- UMR8161 CNRS, Université de Lille, Institut Pasteur de Lille, Institut de Biologie de Lille, 1 Rue Calmette, 59021 Lille CEDEX, France
| | - Randy J Seeley
- Department of Internal Medicine, University of Cincinnati, United States
| | - Yvonne M Ulrich-Lai
- Department of Psychiatry and Behavioral Neuroscience, University of Cincinnati, 2170 East Galbraith Road, Cincinnati, OH 45237, United States
| |
Collapse
|
42
|
Hartanto FK, Karen-Ng LP, Vincent-Chong VK, Ismail SM, Mustafa WMW, Abraham MT, Tay KK, Zain RB. KRT13, FAIM2 and CYP2W1 mRNA Expression in Oral Squamous Cell Carcinoma Patients with Risk Habits. Asian Pac J Cancer Prev 2015; 16:953-8. [DOI: 10.7314/apjcp.2015.16.3.953] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
|
43
|
Abstract
Modified RNA molecules have recently been shown to regulate nervous system functions. This mini-review and associated mini-symposium provide an overview of the types and known functions of novel modified RNAs in the nervous system, including covalently modified RNAs, edited RNAs, and circular RNAs. We discuss basic molecular mechanisms involving RNA modifications as well as the impact of modified RNAs and their regulation on neuronal processes and disorders, including neural fate specification, intellectual disability, neurodegeneration, dopamine neuron function, and substance use disorders.
Collapse
|
44
|
van Gestel MA, Sanders LE, de Jong JW, Luijendijk MCM, Adan RAH. FTO knockdown in rat ventromedial hypothalamus does not affect energy balance. Physiol Rep 2014; 2:2/12/e12152. [PMID: 25501432 PMCID: PMC4332191 DOI: 10.14814/phy2.12152] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Single nucleotide polymorphisms (SNPs) clustered in the first intron of the fat mass and obesity‐associated (FTO) gene has been associated with obesity. FTO expression is ubiquitous, with particularly high levels in the hypothalamic area of the brain. To investigate the region‐specific role of FTO, AAV technology was applied to knockdown FTO in the ventromedial hypothalamus (VMH). No effect of FTO knockdown was observed on bodyweight or parameters of energy balance. Animals were exposed twice to an overnight fast, followed by a high‐fat high‐sucrose (HFHS) diet for 1 week. FTO knockdown did not result in a different response to the diets. A region‐specific role for FTO in the VMH in the regulation of energy balance could not be found. Knocking down expression of the obesity‐associated gene FTO in rat ventromedial hypothalamus did not affect energy balance.
Collapse
Affiliation(s)
- Margriet A van Gestel
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Loek E Sanders
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Johannes W de Jong
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mieneke C M Luijendijk
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Roger A H Adan
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
45
|
Boender AJ, van Gestel MA, Garner KM, Luijendijk MCM, Adan RAH. The obesity-associated gene Negr1 regulates aspects of energy balance in rat hypothalamic areas. Physiol Rep 2014; 2:2/7/e12083. [PMID: 25077509 PMCID: PMC4187548 DOI: 10.14814/phy2.12083] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Neural growth regulator 1 (Negr1) is among the first common variants that have been associated with the regulation of body mass index. Using AAV technology directed to manipulate Negr1 expression in vivo, we find that decreased expression of Negr1 in periventricular hypothalamic areas leads to increases in body weight, presumably via increased food intake. Moreover, we observed that both increased and decreased levels of Negr1 lead to reduced locomotor activity and body temperature. In sum, our results provide further support for a role of hypothalamic expressed Negr1 in the regulation of energy balance.
Collapse
Affiliation(s)
- Arjen J Boender
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Margriet A van Gestel
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Keith M Garner
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Mieneke C M Luijendijk
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Roger A H Adan
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
46
|
AAV-mediated gene transfer of the obesity-associated gene Etv5 in rat midbrain does not affect energy balance or motivated behavior. PLoS One 2014; 9:e94159. [PMID: 24710089 PMCID: PMC3978017 DOI: 10.1371/journal.pone.0094159] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 03/10/2014] [Indexed: 11/19/2022] Open
Abstract
Several genome-wide association studies have implicated the transcription factor E-twenty- six version 5 (Etv5) in the regulation of body mass index. Further substantiating the role of Etv5 in feeding behavior are the findings that targeted disruption of Etv5 in mice leads to decreased body weight gain and that expression of Etv5 is decreased in the ventral tegmental area and substantia nigra pars compacta (VTA/SNpc) after food restriction. As Etv5 has been suggested to influence dopaminergic neurotransmission by driving the expression of genes that are responsible for the synthesis and release of dopamine, we investigated if expression levels of Etv5 are dependent on nutritional state and subsequently influence the expression levels of tyrosine hydroxylase. While it was shown that Etv5 expression in the VTA/SNpc increases after central administration of leptin and that Etv5 was able to drive expression of tyrosine hydroxylase in vitro, AAV-mediated gene transfer of Etv5 into the VTA/SNpc of rats did not alter expression of tyrosine hydroxylase in vivo. Moreover, AAV-mediated gene transfer of Etv5 in the VTA/SNpc did not affect measures of energy balance or performances in a progressive ratio schedule. Thus, these data do not support a role for increased expression of Etv5 in the VTA/SNpc in the regulation of feeding behavior.
Collapse
|
47
|
Corella D, Sorlí JV, González JI, Ortega C, Fitó M, Bulló M, Martínez-González MA, Ros E, Arós F, Lapetra J, Gómez-Gracia E, Serra-Majem L, Ruiz-Gutierrez V, Fiol M, Coltell O, Vinyoles E, Pintó X, Martí A, Saiz C, Ordovás JM, Estruch R. Novel association of the obesity risk-allele near Fas Apoptotic Inhibitory Molecule 2 (FAIM2) gene with heart rate and study of its effects on myocardial infarction in diabetic participants of the PREDIMED trial. Cardiovasc Diabetol 2014; 13:5. [PMID: 24393375 PMCID: PMC3922966 DOI: 10.1186/1475-2840-13-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 12/31/2013] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND The Fas apoptotic pathway has been implicated in type 2 diabetes and cardiovascular disease. Although a polymorphism (rs7138803; G > A) near the Fas apoptotic inhibitory molecule 2 (FAIM2) locus has been related to obesity, its association with other cardiovascular risk factors and disease remains uncertain. METHODS We analyzed the association between the FAIM2-rs7138803 polymorphism and obesity, blood pressure and heart rate in 7,161 participants (48.3% with type 2 diabetes) in the PREDIMED study at baseline. We also explored gene-diet interactions with adherence to the Mediterranean diet (MedDiet) and examined the effects of the polymorphism on cardiovascular disease incidence per diabetes status after a median 4.8-year dietary intervention (MedDiet versus control group) follow-up. RESULTS We replicated the association between the FAIM2-rs7138803 polymorphism and greater obesity risk (OR: 1.08; 95% CI: 1.01-1.16; P = 0.011; per-A allele). Moreover, we detected novel associations of this polymorphism with higher diastolic blood pressure (DBP) and heart rate at baseline (B = 1.07; 95% CI: 0.97-1.28 bmp in AA vs G-carriers for the whole population), that remained statistically significant even after adjustment for body mass index (P = 0.012) and correction for multiple comparisons. This association was greater and statistically significant in type-2 diabetic subjects (B = 1.44: 95% CI: 0.23-2.56 bmp; P = 0.010 for AA versus G-carriers). Likewise, these findings were also observed longitudinally over 5-year follow-up. Nevertheless, we found no statistically significant gene-diet interactions with MedDiet for this trait. On analyzing myocardial infarction risk, we detected a nominally significant (P = 0.041) association in type-2 diabetic subjects (HR: 1.86; 95% CI:1.03-3.37 for AA versus G-carriers), although this association did not remain statistically significant following correction for multiple comparisons. CONCLUSIONS We confirmed the FAIM2-rs7138803 relationship with obesity and identified novel and consistent associations with heart rate in particular in type 2 diabetic subjects. Furthermore, our results suggest a possible association of this polymorphism with higher myocardial infarction risk in type-2 diabetic subjects, although this result needs to be replicated as it could represent a false positive.
Collapse
Affiliation(s)
- Dolores Corella
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, Valencia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Genetic and Molecular Epidemiology Unit, Valencia University, Blasco Ibañez, 15, 46010 Valencia, Spain
| | - Jose V Sorlí
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, Valencia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - José I González
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, Valencia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Carolina Ortega
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, Valencia, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Montserrat Fitó
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Cardiovascula Risk and Nutrition Research Group, Institut Hospital del Mar d’Investigacions Mèdiques (IMIM), Barcelona, Spain
| | - Monica Bulló
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Human Nutrition Unit, Faculty of Medicine, IISPV, University Rovira i Virgili, Reus, Spain
| | - Miguel Angel Martínez-González
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Department of Preventive Medicine and Public Health, School of Medicine, University of Navarra, Pamplona, Spain
| | - Emilio Ros
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Lipid Clinic, Endocrinology and Nutrition Service, Institut d’Investigacions Biomèdiques August Pi Sunyer (IDIBAPS), Hospital Clinic, Barcelona, Spain
| | - Fernando Arós
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Department of Cardiology, Araba University Hospital, Vitoria, Spain
| | - José Lapetra
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Department of Family Medicine, Primary Care Division of Sevilla, San Pablo Health Center, Sevilla, Spain
| | - Enrique Gómez-Gracia
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Department of Epidemiology, School of Medicine, University of Malaga, Malaga, Spain
| | - Lluís Serra-Majem
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Department of Clinical Sciences, University of Las Palmas de Gran Canaria, Las Palmas de Gran Canaria, Spain
| | - Valentina Ruiz-Gutierrez
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Instituto de la Grasa, Consejo Superior de Investigaciones Científicas, Sevilla, Spain
| | - Miquel Fiol
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- University Institute for Health Sciences Investigation, Hospital Son Dureta, Palma de Mallorca, Spain
| | - Oscar Coltell
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Department of Computer Languages and Systems, School of Technology and Experimental Sciences, Jaume I University, Castellón, Spain
| | - Ernest Vinyoles
- Primary Care Division, Catalan Institute of Health, Barcelona, Spain
| | - Xavier Pintó
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Lipids and Vascular Risk Unit, Internal Medicine, Hospital Universitario de Bellvitge, Hospitalet de Llobregat, Barcelona, Spain
| | - Amelia Martí
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Department of Nutrition and Physiology, Faculty of Pharmacy, University of Navarra, Pamplona, Spain
| | - Carmen Saiz
- Department of Preventive Medicine and Public Health, School of Medicine, University of Valencia, Valencia, Spain
| | - José M Ordovás
- Department of Cardiovascular Epidemiology and Population Genetics, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- IMDEA Alimentación, Madrid, Spain
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Ramón Estruch
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Department of Internal Medicine, Hospital Clinic, IDIBAPS, Barcelona, Spain
| |
Collapse
|
48
|
Heni M, Kullmann S, Veit R, Ketterer C, Frank S, Machicao F, Staiger H, Häring HU, Preissl H, Fritsche A. Variation in the obesity risk gene FTO determines the postprandial cerebral processing of food stimuli in the prefrontal cortex. Mol Metab 2013; 3:109-13. [PMID: 24634816 DOI: 10.1016/j.molmet.2013.11.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 11/18/2013] [Accepted: 11/23/2013] [Indexed: 01/08/2023] Open
Abstract
Variation in FTO is the strongest genetic determinant of body weight and has recently been linked with impaired neural processing of food stimuli. However, whether this brain-expressed gene affects neuronal processing of food-related stimuli after ingestion is still poorly understood. In this study, twenty-four participants were examined before, 30 and 120 min after ingesting 75 g of glucose solution or water on two separate days. Functional magnetic resonance imaging (fMRI) during visual food presentation was performed. All participants were genotyped for FTO SNP rs8050136. We detected significant differences between FTO genotypes in the prefrontal cortex 30 min post-glucose load in BOLD-response to food pictures (p=0.0017), while no differences were detected in response to water ingestion or 120 min post-glucose load. Since the prefrontal cortex plays a major role in the inhibitory control of eating, we propose that reduced postprandial activity in FTO risk allele carriers contributes to overeating and obesity.
Collapse
Affiliation(s)
- Martin Heni
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Eberhard Karls University Tübingen, Tübingen, Germany ; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany ; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Stephanie Kullmann
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany ; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany ; fMEG Center, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Ralf Veit
- fMEG Center, Eberhard Karls University Tübingen, Tübingen, Germany ; Institute of Medical Psychology and Behavioral Neurobiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Caroline Ketterer
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Eberhard Karls University Tübingen, Tübingen, Germany ; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany ; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Sabine Frank
- fMEG Center, Eberhard Karls University Tübingen, Tübingen, Germany ; Institute of Medical Psychology and Behavioral Neurobiology, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Fausto Machicao
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany ; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Harald Staiger
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Eberhard Karls University Tübingen, Tübingen, Germany ; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany ; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Hans-Ulrich Häring
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Eberhard Karls University Tübingen, Tübingen, Germany ; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany ; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| | - Hubert Preissl
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany ; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany ; fMEG Center, Eberhard Karls University Tübingen, Tübingen, Germany
| | - Andreas Fritsche
- Department of Internal Medicine, Division of Endocrinology, Diabetology, Angiology, Nephrology and Clinical Chemistry, Eberhard Karls University Tübingen, Tübingen, Germany ; Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany ; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany
| |
Collapse
|
49
|
Speakman JR. Functional analysis of seven genes linked to body mass index and adiposity by genome-wide association studies: a review. Hum Hered 2013; 75:57-79. [PMID: 24081222 DOI: 10.1159/000353585] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Genome-wide association studies (GWAS) have identified a total of about 40 single nucleotide polymorphisms (SNPs) that show significant linkage to body mass index, a widely utilised surrogate measure of adiposity. However, only 8 of these associations have been confirmed by follow-up GWAS using more sophisticated measures of adiposity (computed tomography). Among these 8, there is a SNP close to the gene FTO which has been the subject of considerable work to diagnose its function. The remaining 7 SNPs are adjacent to, or within, the genes NEGR1, TMEM18, ETV5, FLJ35779, LINGO2, SH2B1 and GIPR, most of which are less well studied than FTO, particularly in the context of obesity. This article reviews the available data on the functions of these genes, including information gleaned from studies in humans and animal models. At present, we have virtually no information on the putative mechanism associating the genes FLJ35779 and LINGO2 to obesity. All of these genes are expressed in the brain, and for 2 of them (SH2B1 and GIPR), a direct link to the appetite regulation system is known. SH2B1 is an enhancer of intracellular signalling in the JAK-STAT pathway, and GIPR is the receptor for an appetite-linked hormone (GIP) produced by the alimentary tract. NEGR1, ETV5 and SH2B1 all have suggested roles in neurite outgrowth, and hence SNPs adjacent to these genes may affect development of the energy balance circuitry. Although the genes have central patterns of gene expression, implying a central neuronal connection to energy balance, for at least 4 of them (NEGR1, TMEM18, SH2B1 and GIPR), there are also significant peripheral functions related to adipose tissue biology. These functions may contribute to their effects on the obese phenotype.
Collapse
Affiliation(s)
- John R Speakman
- Key State Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, PR China; Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
50
|
Karra E, O'Daly OG, Choudhury AI, Yousseif A, Millership S, Neary MT, Scott WR, Chandarana K, Manning S, Hess ME, Iwakura H, Akamizu T, Millet Q, Gelegen C, Drew ME, Rahman S, Emmanuel JJ, Williams SCR, Rüther UU, Brüning JC, Withers DJ, Zelaya FO, Batterham RL. A link between FTO, ghrelin, and impaired brain food-cue responsivity. J Clin Invest 2013; 123:3539-51. [PMID: 23867619 DOI: 10.1172/jci44403] [Citation(s) in RCA: 275] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Accepted: 05/17/2013] [Indexed: 12/15/2022] Open
Abstract
Polymorphisms in the fat mass and obesity-associated gene (FTO) are associated with human obesity and obesity-prone behaviors, including increased food intake and a preference for energy-dense foods. FTO demethylates N6-methyladenosine, a potential regulatory RNA modification, but the mechanisms by which FTO predisposes humans to obesity remain unclear. In adiposity-matched, normal-weight humans, we showed that subjects homozygous for the FTO "obesity-risk" rs9939609 A allele have dysregulated circulating levels of the orexigenic hormone acyl-ghrelin and attenuated postprandial appetite reduction. Using functional MRI (fMRI) in normal-weight AA and TT humans, we found that the FTO genotype modulates the neural responses to food images in homeostatic and brain reward regions. Furthermore, AA and TT subjects exhibited divergent neural responsiveness to circulating acyl-ghrelin within brain regions that regulate appetite, reward processing, and incentive motivation. In cell models, FTO overexpression reduced ghrelin mRNA N6-methyladenosine methylation, concomitantly increasing ghrelin mRNA and peptide levels. Furthermore, peripheral blood cells from AA human subjects exhibited increased FTO mRNA, reduced ghrelin mRNA N6-methyladenosine methylation, and increased ghrelin mRNA abundance compared with TT subjects. Our findings show that FTO regulates ghrelin, a key mediator of ingestive behavior, and offer insight into how FTO obesity-risk alleles predispose to increased energy intake and obesity in humans.
Collapse
Affiliation(s)
- Efthimia Karra
- Centre for Obesity Research, University College London, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|