1
|
Westerman KE, Walker ME, Gaynor SM, Wessel J, DiCorpo D, Ma J, Alonso A, Aslibekyan S, Baldridge AS, Bertoni AG, Biggs ML, Brody JA, Chen YDI, Dupuis J, Goodarzi MO, Guo X, Hasbani NR, Heath A, Hidalgo B, Irvin MR, Johnson WC, Kalyani RR, Lange L, Lemaitre RN, Liu CT, Liu S, Moon JY, Nassir R, Pankow JS, Pettinger M, Raffield LM, Rasmussen-Torvik LJ, Selvin E, Senn MK, Shadyab AH, Smith AV, Smith NL, Steffen L, Talegakwar S, Taylor KD, de Vries PS, Wilson JG, Wood AC, Yanek LR, Yao J, Zheng Y, Boerwinkle E, Morrison AC, Fornage M, Russell TP, Psaty BM, Levy D, Heard-Costa NL, Ramachandran VS, Mathias RA, Arnett DK, Kaplan R, North KE, Correa A, Carson A, Rotter JI, Rich SS, Manson JE, Reiner AP, Kooperberg C, Florez JC, Meigs JB, Merino J, Tobias DK, Chen H, Manning AK. Investigating Gene-Diet Interactions Impacting the Association Between Macronutrient Intake and Glycemic Traits. Diabetes 2023; 72:653-665. [PMID: 36791419 PMCID: PMC10130485 DOI: 10.2337/db22-0851] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Accepted: 02/02/2023] [Indexed: 02/17/2023]
Abstract
Few studies have demonstrated reproducible gene-diet interactions (GDIs) impacting metabolic disease risk factors, likely due in part to measurement error in dietary intake estimation and insufficient capture of rare genetic variation. We aimed to identify GDIs across the genetic frequency spectrum impacting the macronutrient-glycemia relationship in genetically and culturally diverse cohorts. We analyzed 33,187 participants free of diabetes from 10 National Heart, Lung, and Blood Institute Trans-Omics for Precision Medicine program cohorts with whole-genome sequencing, self-reported diet, and glycemic trait data. We fit cohort-specific, multivariable-adjusted linear mixed models for the effect of diet, modeled as an isocaloric substitution of carbohydrate for fat, and its interactions with common and rare variants genome-wide. In main effect meta-analyses, participants consuming more carbohydrate had modestly lower glycemic trait values (e.g., for glycated hemoglobin [HbA1c], -0.013% HbA1c/250 kcal substitution). In GDI meta-analyses, a common African ancestry-enriched variant (rs79762542) reached study-wide significance and replicated in the UK Biobank cohort, indicating a negative carbohydrate-HbA1c association among major allele homozygotes only. Simulations revealed that >150,000 samples may be necessary to identify similar macronutrient GDIs under realistic assumptions about effect size and measurement error. These results generate hypotheses for further exploration of modifiable metabolic disease risk in additional cohorts with African ancestry. ARTICLE HIGHLIGHTS We aimed to identify genetic modifiers of the dietary macronutrient-glycemia relationship using whole-genome sequence data from 10 Trans-Omics for Precision Medicine program cohorts. Substitution models indicated a modest reduction in glycemia associated with an increase in dietary carbohydrate at the expense of fat. Genome-wide interaction analysis identified one African ancestry-enriched variant near the FRAS1 gene that may interact with macronutrient intake to influence hemoglobin A1c. Simulation-based power calculations accounting for measurement error suggested that substantially larger sample sizes may be necessary to discover further gene-macronutrient interactions.
Collapse
Affiliation(s)
- Kenneth E. Westerman
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA
| | - Maura E. Walker
- Department of Medicine, Section of Preventive Medicine, Boston University School of Medicine, Boston, MA
- Department of Health Sciences, Sargent College of Health and Rehabilitation Sciences, Boston University, Boston, MA
| | - Sheila M. Gaynor
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Jennifer Wessel
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indianapolis, IN
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
- Diabetes Translational Research Center, Indiana University, Indianapolis, IN
| | - Daniel DiCorpo
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Jiantao Ma
- Nutrition Epidemiology and Data Science, Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA
| | - Alvaro Alonso
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | | | - Abigail S. Baldridge
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Alain G. Bertoni
- Department of Epidemiology and Prevention, Wake Forest School of Medicine, Winston-Salem, NC
| | - Mary L. Biggs
- Department of Biostatistics, University of Washington, Seattle, WA
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA
| | - Jennifer A. Brody
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
| | - Yii-Der Ida Chen
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Joseé Dupuis
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
| | - Mark O. Goodarzi
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Natalie R. Hasbani
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Adam Heath
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Bertha Hidalgo
- School of Public Health, University of Alabama at Birmingham, Birmingham, AL
| | - Marguerite R. Irvin
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL
| | - W. Craig Johnson
- Department of Biostatistics, University of Washington, Seattle, WA
| | - Rita R. Kalyani
- GeneSTAR Research Program, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Leslie Lange
- Department of Medicine, Anschutz Medical Campus, University of Colorado, Aurora, CO
| | - Rozenn N. Lemaitre
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA
- Department of Internal Medicine, University of Washington, Seattle, WA
| | - Ching-Ti Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA
- National Heart, Lung, and Blood Institute and Boston University’s Framingham Heart Study, Framingham, MA
- Evans Department of Medicine, Section of Preventive Medicine and Epidemiology, Boston University School of Medicine, Boston, MA
- Evans Department of Medicine, Whitaker Cardiovascular Institute and Cardiology Section, Boston University School of Medicine, Boston, MA
| | - Simin Liu
- Center for Global Cardiometabolic Health, Boston, MA
| | - Jee-Young Moon
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Rami Nassir
- Department of Pathology, School of Medicine, Umm Al-Qura University, Mecca, Saudi Arabia
| | - James S. Pankow
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Mary Pettinger
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Laura M. Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | | | - Elizabeth Selvin
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Mackenzie K. Senn
- USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX
| | - Aladdin H. Shadyab
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla, CA
| | - Albert V. Smith
- Department of Biostatistics, University of Michigan, Ann Arbor, MI
| | - Nicholas L. Smith
- Department of Epidemiology, University of Washington, Seattle, WA
- Kaiser Permanente Washington Health Research Institute, Seattle, WA
- Department of Veterans Affairs Office of Research and Development, Seattle Epidemiologic Research and Information Center, Seattle, WA
| | - Lyn Steffen
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, Minneapolis, MN
| | - Sameera Talegakwar
- Department of Exercise and Nutrition Sciences, Milken Institute School of Public Health, The George Washington University, Washington, DC
| | - Kent D. Taylor
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Paul S. de Vries
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - James G. Wilson
- Division of Cardiovascular Medicine, Beth Israel Deaconess Medical Center, Boston, MA
| | - Alexis C. Wood
- USDA/ARS Children’s Nutrition Research Center, Baylor College of Medicine, Houston, TX
| | - Lisa R. Yanek
- GeneSTAR Research Program, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Yinan Zheng
- Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL
| | - Eric Boerwinkle
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX
| | - Alanna C. Morrison
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Miriam Fornage
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Tracy P. Russell
- Department of Pathology and Laboratory Medicine, University of Vermont Larner College of Medicine, Burlington, VT
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, University of Washington, Seattle, WA
- Department of Medicine, University of Washington, Seattle, WA
- Department of Epidemiology, University of Washington, Seattle, WA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA
| | - Daniel Levy
- National Heart, Lung, and Blood Institute and Boston University’s Framingham Heart Study, Framingham, MA
- Population Sciences Branch, Division of Intramural Research, National Heart, Lung, and Blood Institute, Bethesda, MD
| | - Nancy L. Heard-Costa
- National Heart, Lung, and Blood Institute and Boston University’s Framingham Heart Study, Framingham, MA
- Department of Neurology, Boston University School of Medicine, Boston, MA
| | - Vasan S. Ramachandran
- National Heart, Lung, and Blood Institute and Boston University’s Framingham Heart Study, Framingham, MA
- Evans Department of Medicine, Section of Preventive Medicine and Epidemiology, Boston University School of Medicine, Boston, MA
- Evans Department of Medicine, Whitaker Cardiovascular Institute and Cardiology Section, Boston University School of Medicine, Boston, MA
| | - Rasika A. Mathias
- GeneSTAR Research Program, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Donna K. Arnett
- College of Public Health, University of Kentucky, Lexington, KY
| | - Robert Kaplan
- Clinical Excellence Research Center, School of Medicine, Stanford University, Stanford, CA
| | - Kari E. North
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Adolfo Correa
- Department of Population Health Science, University of Mississippi Medical Center, Jackson, MS
| | - April Carson
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Stephen S. Rich
- Center for Public Health Genomics, Department of Public Health Sciences, University of Virginia, Charlottesville, VA
| | | | | | - Charles Kooperberg
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Jose C. Florez
- Department of Medicine, Harvard Medical School, Boston, MA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA
- Diabetes Unit, Massachusetts General Hospital, Boston, MA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
| | - James B. Meigs
- Department of Medicine, Harvard Medical School, Boston, MA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA
- Division of General Internal Medicine, Massachusetts General Hospital, Boston, MA
| | - Jordi Merino
- Department of Medicine, Harvard Medical School, Boston, MA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA
- Diabetes Unit, Massachusetts General Hospital, Boston, MA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA
| | - Deirdre K. Tobias
- Division of Preventive Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Han Chen
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
- Center for Precision Health, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Alisa K. Manning
- Clinical and Translational Epidemiology Unit, Mongan Institute, Massachusetts General Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
- Programs in Metabolism and Medical and Population Genetics, Broad Institute of MIT and Harvard, Boston, MA
| |
Collapse
|
2
|
Overweight and obesity in preschool children in Turkey: A multilevel analysis. J Biosoc Sci 2023; 55:344-366. [PMID: 35086578 DOI: 10.1017/s0021932022000025] [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: 11/06/2022]
Abstract
Childhood obesity/overweight is a worldwide concern and its prevalence is increasing in many countries. The first aim of this study is to analyse the trends in overweight and obesity among children under the age of five in Turkey based on the new World Health Organization (WHO) standards, using data from the 'five-round of the Turkey Demographic and Health Surveys' (TDHSs). The second aim is to examine whether or not the maternal/household and individual-level factors are associated with overweight/obesity using TDHS 2003, 2008, and 2013 datasets. A total sample of 14,231 children under the age of five were extracted from the TDHS in 1993, 1998, 2003, 2008, and 2013 to determine the prevalence of the trend. Pooled data from 8,812 children were included in the analysis to examine factors associated with overweight/obesity. Taking into account the clustered data structure, multilevel logistic regression models were utilised. In 1993, 1998, 2003, 2008, and 2013 the prevalence of overweight children was 5.3%, 4.9%, 10.0%, 11% and 11.6%, respectively. The factors that were independently associated with overweight/obesity were as follows: living in single-parent households (adjusted odds ratio (aOR) = 2.27, 95%CI = 1.21-4.26), compared to living in dual-parent households; having an obese mother (aOR = 4.25, 95%CI = 1.73-10.44), overweight mother (aOR = 3.15, 95%CI = 1.29-7.69), and a normal-weight mother (aOR = 2.70, 95%CI = 1.11-6.59) compared to having an underweight mother; being aged between 13-24 months (aOR = 1.72, 95%CI = 1.30 to 2.27), compared to being aged 0-12 months; male gender (aOR = 1.30, 95%CI = 1.11 to 1.53); being stunted (aOR = 2.18, 95%CI = 1.74 to 2.73); high birth weight (aOR = 1.55, 95%CI = 1.08 to 2.23) compared to low birth weight. In addition, overweight was higher in children of mothers who had completed primary school (aOR = 1.21, 95%CI = 1.01 to 1.59) than children of mothers who had not completed primary school. These findings reveal that, over the years, there has been a substantial increase in obesity/overweight among children which demonstrates the importance of evaluating the overweight indicators at the maternal/household level.
Collapse
|
3
|
Banik S. Genetic, Epigenetic, and Molecular Biology of Obesity: From Pathology to Therapeutics the Way Forward. JOURNAL OF THE ASSOCIATION OF PHYSICIANS OF INDIA 2022; 70:11-12. [DOI: 10.5005/japi-11001-0080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
4
|
Interaction between Apo A-II -265T > C polymorphism and dietary total antioxidant capacity on some oxidative stress and inflammatory markers in patients with type 2 diabetes mellitus. Br J Nutr 2022; 128:13-29. [PMID: 34372957 DOI: 10.1017/s0007114521002993] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
This work aims to examine the interaction between apo A2 (Apo A-II) -265T > C SNP and dietary total antioxidant capacity (DTAC) on inflammation and oxidative stress in patients with type 2 diabetes mellitus. The present cross-sectional study included 180 patients (35-65 years) with identified Apo A-II genotype. Dietary intakes were assessed by a FFQ. DTAC was computed using the international databases. IL-18 (IL18), high-sensitivity C-reactive protein (hs-CRP), pentraxin (PTX3), serum total antioxidant capacity (TAC), superoxide dismutase (SOD) activity and 8-isoprostaneF2α (PGF2α) markers were obtained according to standard protocols. General linear model was used to evaluate the interaction. The interaction of gene and DTAC (PFRAP = 0·039 and PORAC = 0·042) on PGF2α level was significant after adjusting for confounders. A significant interaction was observed on IL18 level (PORAC = 0·018 and PFRAP = 0·048) and SOD (PTEAC = 0·037) in obese patients. Among patients whose DTAC was higher than the median intake, the levels of hs-CRP and PGF2α were significantly higher only in individuals with CC genotype. Serum TAC (PFRAP = 0·030, PORAC = 0·049) and SOD were significantly lower in the CC genotype. There was a favourable relationship between the high-DTAC and SOD (obese: PTEAC = 0·034, non-obese: PFRAP = 0·001, PTRAP < 0·0001, PTEAC = 0·003 and PORAC = 0·001) and PGF2α (non-obese: PORAC = 0·024) in T-allele carriers. The rs5082 SNP interacts with DTAC to influence several cardiometabolic risk factors. Also, we found dietary recommendations for antioxidant-rich foods intake might be useful in the prevention of diabetes complications in the T carrier more effectively than the CC genotype. Future large studies are required to confirm these results.
Collapse
|
5
|
Abstract
Obesity is as a global health problem due to its interaction with complex chronic disorders such as cardiovascular disorders, type 2 diabetes mellitus (T2DM) and cancer. Despite the fact that pathogenesis of obesity is not yet clearly understood, it is associated with a combination of psychological, environmental and various genetic factors. Here, employing a case-control design, we aimed to examine the effects of the GHRL c.152C>T (p.Arg51Gln) (rs34911341) and c.214G>T (p.Leu72Met) (rs696217) markers on susceptibility to obesity in a Turkish-Cypriot population, as well as to evaluate whether these markers affect biochemical parameters and show their putative functional consequences. This study involved 211 Turkish-Cypriot subjects (106 obese and 95 non obese). Genotyping for the GHRL gene polymorphisms was performed by polymerase chain reaction-restriction fragment length polymorphism (PCR-RFLP) analysis. Our results indicate that the GHRL Leu72Met polymorphism was found to be significantly higher in obese patients, with respect to genotypic (p = 0.0012) and allelic (p = 0.0005) frequencies. Strikingly, the rs696217 GT genotype (heterozygous) had significantly lower serum high-density lipoprotein cholesterol (HDL-C) (p = 0.015) than GG (wild type) genotypes. Overall, Leu72Met susceptibility variant may be considered as risk and crucial marker for both obesity and cholesterol metabolism in the community of Turkish-Cypriots. Thus, the dual effect of the GHRL gene Leu72Met variant may be used for clinical diagnosis.
Collapse
|
6
|
Ghanemi A, Yoshioka M, St-Amand J. Trefoil Factor Family Member 2: From a High-Fat-Induced Gene to a Potential Obesity Therapy Target. Metabolites 2021; 11:metabo11080536. [PMID: 34436477 PMCID: PMC8401738 DOI: 10.3390/metabo11080536] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 12/11/2022] Open
Abstract
Obesity has its epidemiological patterns continuously increasing. With controlling both diet and exercise being the main approaches to manage the energy metabolism balance, a high-fat (HF) diet is of particular importance. Indeed, lipids have a low satiety potential but a high caloric density. Thus, focusing on pharmacologically targetable pathways remains an approach with promising therapeutic potential. Within this context, trefoil factor family member 2 (Tff2) has been characterized as specifically induced by HF diet rather than low-fat diet. TFF2 has also been linked to diverse neurological mechanisms and metabolic patterns suggesting its role in energy balance. The hypothesis is that TFF2 would be a HF diet-induced signal that regulates metabolism with a focus on lipids. Within this review, we put the spotlight on key findings highlighting this line of thought. Importantly, the hypothetical mechanisms pointed highlight TFF2 as an important contributor to obesity development via increasing lipids intestinal absorption and anabolism. Therefore, an outlook for future experimental activities and evaluation of the therapeutic potential of TFF2 inhibition is given. Indeed, its knockdown or downregulation would contribute to an antiobesity phenotype. We believe this work represents an addition to our understanding of the lipidic molecular implications in obesity, which will contribute to develop therapies aiming to manage the lipidic metabolic pathways including the absorption, storage and metabolism via targeting TFF2-related pathways. We briefly discuss important relevant concepts for both basic and clinical researchers.
Collapse
Affiliation(s)
- Abdelaziz Ghanemi
- Functional Genomics Laboratory, CREMI, Québec Genome Center, CHUL-CHU de Québec Research Center, Quebec, QC G1V 4G2, Canada; (A.G.); (M.Y.)
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada
| | - Mayumi Yoshioka
- Functional Genomics Laboratory, CREMI, Québec Genome Center, CHUL-CHU de Québec Research Center, Quebec, QC G1V 4G2, Canada; (A.G.); (M.Y.)
| | - Jonny St-Amand
- Functional Genomics Laboratory, CREMI, Québec Genome Center, CHUL-CHU de Québec Research Center, Quebec, QC G1V 4G2, Canada; (A.G.); (M.Y.)
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec, QC G1V 0A6, Canada
- Correspondence:
| |
Collapse
|
7
|
The influence of polymorphisms of fat mass and obesity (FTO, rs9939609) and vitamin D receptor (VDR, BsmI, TaqI, ApaI, FokI) genes on weight loss by diet and exercise interventions in non-diabetic overweight/obese Asian Indians in North India. Eur J Clin Nutr 2020; 74:604-612. [DOI: 10.1038/s41430-020-0560-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 01/09/2020] [Accepted: 01/14/2020] [Indexed: 01/19/2023]
|
8
|
Jiang L, Audouze K, Romero Herrera JA, Ängquist LH, Kjærulff SK, Izarzugaza JM, Tjønneland A, Halkjær J, Overvad K, Sørensen TI, Brunak S. Conflicting associations between dietary patterns and changes of anthropometric traits across subgroups of middle-aged women and men. Clin Nutr 2020; 39:265-275. [DOI: 10.1016/j.clnu.2019.02.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 12/20/2018] [Accepted: 02/04/2019] [Indexed: 01/08/2023]
|
9
|
Kim TJ, Makowski AC, von dem Knesebeck O. Obesity stigma in Germany and the United States - Results of population surveys. PLoS One 2019; 14:e0221214. [PMID: 31430315 PMCID: PMC6701774 DOI: 10.1371/journal.pone.0221214] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 08/02/2019] [Indexed: 12/05/2022] Open
Abstract
Introduction Over the past decades, obesity stigma has become a substantial public health issue as studies have highlighted its negative consequences for mental and physical health. However, comparative studies are scarce. In this cross-national study, we focus on the following research questions: (1) Are there differences in the magnitude of public obesity stigma between Germany and the United States (US), and (2) are there differences in the associations of sociodemographic as well as experience (i.e. former obesity experience) and contact-related (i.e. contact to a person with obesity) factors with public obesity stigma between these two countries? Material and methods National telephone surveys in Germany and the United States were conducted (total sample = 2,802) by using vignettes for the description of a person with obesity. Fat Phobia Scale, negative reactions, and desire for social distance were assessed as components of public obesity stigma. All three stigma components were examined with multilevel linear regression analyses. Results Overall, results show that public obesity stigma is significantly more pronounced in the US than in Germany. Relationships between obesity stigma and sociodemographic as well as experience and contact-related factors remain rather inconclusive, though, in general, stronger associations are shown in the US. Conclusions Contrary to the normalization hypothesis, findings indicate that a comparatively high prevalence of obesity like in the US is associated with a higher level of obesity stigma.
Collapse
Affiliation(s)
- Tae Jun Kim
- Department of Medical Sociology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- * E-mail:
| | - Anna Christin Makowski
- Department of Medical Sociology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Olaf von dem Knesebeck
- Department of Medical Sociology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
10
|
Gene-Environment Interactions on Body Fat Distribution. Int J Mol Sci 2019; 20:ijms20153690. [PMID: 31357654 PMCID: PMC6696304 DOI: 10.3390/ijms20153690] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Revised: 07/18/2019] [Accepted: 07/19/2019] [Indexed: 02/08/2023] Open
Abstract
The prevalence of obesity has been increasing markedly in the U.S. and worldwide in the past decades; and notably, the obese populations are signified by not only the overall elevated adiposity but also particularly harmful accumulation of body fat in the central region of the body, namely, abdominal obesity. The profound shift from “traditional” to “obesogenic” environments, principally featured by the abundance of palatable, energy-dense diet, reduced physical activity, and prolonged sedentary time, promotes the obesity epidemics and detrimental body fat distribution. Recent advances in genomics studies shed light on the genetic basis of obesity and body fat distribution. In addition, growing evidence from investigations in large cohorts and clinical trials has lent support to interactions between genetic variations and environmental factors, e.g., diet and lifestyle factors, in relation to obesity and body fat distribution. This review summarizes the recent discoveries from observational studies and randomized clinical trials on the gene–environment interactions on obesity and body fat distribution.
Collapse
|
11
|
Shrestha D, Rahman ML, Workalemahu T, Zhu C, Tekola-Ayele F. Influence of Fetal and Maternal Genetic Susceptibility to Obesity on Birthweight in African Ancestry Populations. Front Genet 2018; 9:511. [PMID: 30450111 PMCID: PMC6224338 DOI: 10.3389/fgene.2018.00511] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 10/12/2018] [Indexed: 02/02/2023] Open
Abstract
Fetal and maternal genetic propensity to obesity can influence birthweight. We investigated the effects of fetal and maternal genetic risk of obesity on birthweight and evaluated whether these genetic influences modify the well-known association between maternal pre-pregnancy body mass index (BMI) and birthweight. In 950 mother-baby pairs of African ancestry, a genetic risk score for adulthood obesity was generated for mothers (mGRS) and their babies (bGRS) as the weighted sum of BMI-increasing alleles of 97 single nucleotide polymorphisms known to be associated with BMI. The median GRS value was used as a cut-off to define high or low bGRS and mGRS. High bGRS was significantly associated with 70 g lower birthweight (95% Confidence Interval [CI] = −127.4 to −12.4) compared to low bGRS. mGRS was positively correlated with birthweight but the association was not significant. mGRS modified the significant birthweight-increasing effect of maternal pre-pregnancy BMI (P-for-interaction = 0.03); among mothers with low mGRS, those who were overweight or obese had 127.7 g heavier babies (95% CI = 27.1 to 228.2) compared to those who had normal weight. In summary, fetal obesity genetic risk loci exert direct influence on birthweight, and maternal loci modify the effect of pre-pregnancy BMI on birthweight.
Collapse
Affiliation(s)
- Deepika Shrestha
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Mohammad L Rahman
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Tsegaselassie Workalemahu
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Chunming Zhu
- Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Fasil Tekola-Ayele
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
12
|
Beckett EL, Jones PR, Veysey M, Lucock M. Nutrigenetics—Personalized Nutrition in the Genetic Age. EXPLORATORY RESEARCH AND HYPOTHESIS IN MEDICINE 2017; 2:1-8. [DOI: 10.14218/erhm.2017.00027] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
13
|
Ye K, Li L, Zhang D, Li Y, Wang HQ, Lai HL, Hu CL. Effect of Maternal Obesity on Fetal Growth and Expression of Placental Fatty Acid Transporters. J Clin Res Pediatr Endocrinol 2017; 9:300-307. [PMID: 28588000 PMCID: PMC5785635 DOI: 10.4274/jcrpe.4510] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
OBJECTIVE To explore the effects of maternal high-fat (HF) diet-induced obesity on fetal growth and the expression of placental nutrient transporters. METHODS Maternal obesity was established in rats by 8 weeks of pre-pregnancy fed HF diet, while rats in the control group were fed normal (CON) diet. Diet-induced obesity (DIO) rats and diet-induced obesity-resistant (DIR) rats were selected according to body weight gain over this period. After copulation, the CON rats were divided into two groups: switched to HF diet (CON-HF group) or maintained on the CON diet (CON-CON group). The DIO rats and DIR rats were maintained on the HF diet throughout pregnancy. Pregnant rats were euthanized at day 21 gestation, fetal and placental weights were recorded, and placental tissue was collected. Reverse transcription-polymerase chain reaction was used to determine mRNA expression of placental nutrient transporters. Protein expression was determined by Western blot. RESULTS Average fetal weight of DIO dams was reduced by 6.9%, and the placentas of CON-HF and DIO dams were significantly heavier than the placentas of CON-CON and DIR dams at day 21 of gestation (p<0.05). The fetal/placental weight ratio of DIO dams was significantly reduced compared with the fetal/placental weight ratio of CON-CON dams (p<0.05). The mRNA expression of GLUT-1 and SNAT-2 were not significantly different between groups. The mRNA and protein expression levels of CD36, FATP-1, and FATP-4 in DIO dams were decreased significantly (p<0.05). CONCLUSION Maternal obesity induced by a HF diet led to intrauterine growth retardation and down-regulated the expression of placental fatty acid transporters.
Collapse
Affiliation(s)
- Kui Ye
- Anhui Medical University School of Public Health, Department of Nutrition and Food Hygiene, Anhui, China
| | - Li Li
- Anhui Medical University School of Public Health, Department of Nutrition and Food Hygiene, Anhui, China
,* Address for Correspondence: Anhui Medical University School of Public Health, Department of Nutrition and Food Hygiene, Anhui, China Phone: +86 551 63869176 E-mail:
| | - Dan Zhang
- Lujiang Center for Disease Control and Prevention, Department of Public Health, Anhui, China
| | - Yi Li
- Anhui Provincial Hospital, Clinic of Clinical Nutrition, Anhui, China
| | - Hai-Qing Wang
- Anhui Medical University School of Public Health, Department of Nutrition and Food Hygiene, Anhui, China
| | - Han-Lin Lai
- Anhui Medical University School of Public Health, Department of Nutrition and Food Hygiene, Anhui, China
| | - Chuan-Lai Hu
- Anhui Medical University School of Public Health, Department of Nutrition and Food Hygiene, Anhui, China
| |
Collapse
|
14
|
Depot-specific inflammation with decreased expression of ATM2 in white adipose tissues induced by high-margarine/lard intake. PLoS One 2017; 12:e0188007. [PMID: 29141038 PMCID: PMC5687764 DOI: 10.1371/journal.pone.0188007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 10/30/2017] [Indexed: 01/21/2023] Open
Abstract
A high-fat diet has been recognized as an important risk factor of obesity, with variable impacts of different fatty acid compositions on the physiological process. To understand the effects of a high-margarine/lard diet, which is a major source of trans fatty acids (TFAs)/ saturated fatty acids (SFAs), elaidic acid as a biomarker of margarine intake was used to screen affected adipokines on mature human adipocytes in vitro. Weaned male Wistar rats were fed a high-fat diet enriched with margarine/lard to generate obesity-prone (OP) and obesity-resistant (OR) models, which were then used to explore the inflammatory responses of depot-specific white adipose tissue. Adiposity, glucose and lipid metabolism parameters and macrophage cell markers were also compared in vivo. In the subcutaneous depot, a high-margarine diet induced elevated IL-6, MCP-1 and XCL1 expression levels in both M-OP and M-OR groups. High-lard diet-fed rats displayed higher protein expression levels of MCP-1 and XCL1 compared with the control group. In the epididymal depot, significantly elevated IL-6 production was observed in M-OP rats, and high-lard diet-fed rats displayed elevated IL-6 and decreased XCL1 expression. In the retroperitoneal depot, a high-margarine diet caused higher IL-6 and MCP-1 expression levels, a high-lard diet caused elevated IL-6 expression in L-OP/L-OR rats, and elevated XCL1 expression was observed only in L-OP rats. In general, CD206 mRNA levels were notably down-regulated by high-fat diet feeding in the above-mentioned depots. CD11c mRNA levels were slightly upregulated in the subcutaneous depot of OP rats fed a high-margarine/lard diet. In the epidydimal depot, higher expression levels of F4/80 and CD206 mRNA were observed only in high-margarine diet-fed OP rats. These results suggest that depot-specific inflammation with decreased expression of adipose tissue anti-inflammatory M2-type (ATM2) macrophages could be induced by high-margarine/lard intake.
Collapse
|
15
|
Gene-nutrient interactions and susceptibility to human obesity. GENES AND NUTRITION 2017; 12:29. [PMID: 29093760 PMCID: PMC5663124 DOI: 10.1186/s12263-017-0581-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 07/04/2017] [Indexed: 12/28/2022]
Abstract
A large number of genome-wide association studies, transferability studies, and candidate gene studies performed in diverse populations around the world have identified gene variants that are associated with common human obesity. The mounting evidence suggests that these obesity gene variants interact with multiple environmental factors and increase susceptibility to this complex metabolic disease. The objective of this review article is to provide concise and updated information on energy balance, heritability of body weight, origins of gene variants, and gene-nutrient interactions in relation to human obesity. It is proposed that knowledge of these related topics will provide valuable insight for future preventative lifestyle intervention using targeted nutritional and medicinal therapies.
Collapse
|
16
|
Celis-Morales CA, Lyall DM, Gray SR, Steell L, Anderson J, Iliodromiti S, Welsh P, Guo Y, Petermann F, Mackay DF, Bailey MES, Pell JP, Gill JMR, Sattar N. Dietary fat and total energy intake modifies the association of genetic profile risk score on obesity: evidence from 48 170 UK Biobank participants. Int J Obes (Lond) 2017; 41:1761-1768. [PMID: 28736445 DOI: 10.1038/ijo.2017.169] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 05/19/2017] [Accepted: 07/05/2017] [Indexed: 01/19/2023]
Abstract
BACKGROUND Obesity is a multifactorial condition influenced by both genetics and lifestyle. The aim of this study was to investigate whether the association between a validated genetic profile risk score for obesity (GPRS-obesity) and body mass index (BMI) or waist circumference (WC) was modified by macronutrient intake in a large general population study. METHODS This study included cross-sectional data from 48 170 white European adults, aged 37-73 years, participating in the UK Biobank. Interactions between GPRS-obesity and macronutrient intake (including total energy, protein, fat, carbohydrate and dietary fibre intake) and its effects on BMI and WC were investigated. RESULTS The 93-single-nucleotide polymorphism (SNP) GPRS was associated with a higher BMI (β: 0.57 kg m-2 per s.d. increase in GPRS (95% confidence interval: 0.53-0.60); P=1.9 × 10-183) independent of major confounding factors. There was a significant interaction between GPRS and total fat intake (P(interaction)=0.007). Among high-fat-intake individuals, BMI was higher by 0.60 (0.52, 0.67) kg m-2 per s.d. increase in GPRS-obesity; the change in BMI with GPRS was lower among low-fat-intake individuals (β: 0.50 (0.44, 0.57) kg m-2). Significant interactions with similar patterns were observed for saturated fat intake (high β: 0.66 (0.59, 0.73) versus low β: 0.49 (0.42, 0.55) kg m-2, P(interaction)=2 × 10-4) and for total energy intake (high β: 0.58 (0.51, 0.64) versus low β: 0.49 (0.42, 0.56) kg m-2, P(interaction)=0.019), but not for protein intake, carbohydrate intake and fibre intake (P(interaction) >0.05). The findings were broadly similar using WC as the outcome. CONCLUSIONS These data suggest that the benefits of reducing the intake of fats and total energy intake may be more important in individuals with high genetic risk for obesity.
Collapse
Affiliation(s)
- C A Celis-Morales
- BHF Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - D M Lyall
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - S R Gray
- BHF Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - L Steell
- BHF Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - J Anderson
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - S Iliodromiti
- BHF Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - P Welsh
- BHF Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Y Guo
- BHF Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - F Petermann
- BHF Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - D F Mackay
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - M E S Bailey
- School of Life Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - J P Pell
- Institute of Health and Wellbeing, University of Glasgow, Glasgow, UK
| | - J M R Gill
- BHF Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - N Sattar
- BHF Cardiovascular Research Centre, Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
17
|
Martínez-Micaelo N, González-Abuín N, Terra X, Ardévol A, Pinent M, Petretto E, Behmoaras J, Blay M. Identification of a nutrient-sensing transcriptional network in monocytes by using inbred rat models on a cafeteria diet. Dis Model Mech 2016; 9:1231-1239. [PMID: 27483348 PMCID: PMC5087837 DOI: 10.1242/dmm.025528] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 07/04/2016] [Indexed: 01/21/2023] Open
Abstract
Obesity has reached pandemic levels worldwide. The current models of diet-induced obesity in rodents use predominantly high-fat based diets that do not take into account the consumption of variety of highly palatable, energy-dense foods that are prevalent in Western society. We and others have shown that the cafeteria (CAF) diet is a robust and reproducible model of human metabolic syndrome with tissue inflammation in the rat. We have previously shown that inbred rat strains such as Wistar Kyoto (WKY) and Lewis (LEW) show different susceptibilities to CAF diets with distinct metabolic and morphometric profiles. Here, we show a difference in plasma MCP-1 levels and investigate the effect of the CAF diet on peripheral blood monocyte transcriptome, as powerful stress-sensing immune cells, in WKY and LEW rats. We found that 75.5% of the differentially expressed transcripts under the CAF diet were upregulated in WKY rats and were functionally related to the activation of the immune response. Using a gene co-expression network constructed from the genes differentially expressed between CAF diet-fed LEW and WKY rats, we identified acyl-CoA synthetase short-chain family member 2 (Acss2) as a hub gene for a nutrient-sensing cluster of transcripts in monocytes. The Acss2 genomic region is significantly enriched for previously established metabolism quantitative trait loci in the rat. Notably, monocyte expression levels of Acss2 significantly correlated with plasma glucose, triglyceride, leptin and non-esterified fatty acid (NEFA) levels as well as morphometric measurements such as body weight and the total fat following feeding with the CAF diet in the rat. These results show the importance of the genetic background in nutritional genomics and identify inbred rat strains as potential models for CAF-diet-induced obesity. Summary: Feeding with a cafeteria diet (CAF) is a reproducible model of human metabolic syndrome in the rat. By using inbred rat models of nutrigenomics, we have studied the effect of CAF on monocyte transcriptome.
Collapse
Affiliation(s)
- Neus Martínez-Micaelo
- Mobiofood Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43003 Tarragona, Spain
| | - Noemi González-Abuín
- Mobiofood Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43003 Tarragona, Spain
| | - Ximena Terra
- Mobiofood Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43003 Tarragona, Spain
| | - Ana Ardévol
- Mobiofood Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43003 Tarragona, Spain
| | - Montserrat Pinent
- Mobiofood Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43003 Tarragona, Spain
| | - Enrico Petretto
- MRC Clinical Sciences Centre, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK Duke-NUS Graduate Medical School Singapore, 8 College Road, Singapore 169857, Republic of Singapore
| | - Jacques Behmoaras
- Centre of Complement and Inflammation Research, Imperial College London, Du Cane Road, London W12 0NN, UK
| | - Mayte Blay
- Mobiofood Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43003 Tarragona, Spain
| |
Collapse
|
18
|
Kowalska K, Olejnik A. Beneficial effects of cranberry in the prevention of obesity and related complications: Metabolic syndrome and diabetes – A review. J Funct Foods 2016. [DOI: 10.1016/j.jff.2015.11.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
|
19
|
Mărginean CO, Bănescu C, Duicu C, Voidăzan S, Mărginean C. Angiotensin-converting enzyme gene insertion/deletion polymorphism in nutritional disorders in children. Eur J Nutr 2015; 54:1245-1254. [PMID: 25416682 DOI: 10.1007/s00394-014-0802-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 11/07/2014] [Indexed: 02/08/2023]
Abstract
PURPOSE The aim of this study was to establish the role of angiotensin-converting enzyme gene insertion/deletion (ACE I/D) polymorphism in determining obesity or undernutrition in a child population in Romania. METHODS We assessed 293 consecutively hospitalized patients in a tertiary emergency pediatric hospital. The patients were divided, according to body mass index (BMI), into three groups: group I, the control group consisting of 126 children, group II patients with undernutrition (85 patients) and group III patients with obesity (82 patients). ACE I/D polymorphisms were performed in all three patient groups, as well as the measuring of anthropometric parameters [middle upper arm circumference (MUAC), tricipital skinfold thickness (TST)]. All patients also underwent paraclinical evaluations (protein and albumin). The cutoffs criteria for moderate undernutrition were: BMI between -2.0 SD and -3.0 SD, severe undernutrition: BMI <-3.0 SD, moderate obesity: BMI between +2.0 SD and +3.0 SD and severe obesity: BMI >+3.0 SD. RESULTS We observed that DD genotype (64.7%) was prevalent in the moderate undernutrition group, while ID (35.3%) and II genotypes were higher in the subgroup of severe undernutrition, with significant correlations in DD and ID genotype groups between BMI and MUAC, protein and albumin (p < 0.0001). In the obese group, we observed significant correlations in DD genotype, between BMI and MUAC (p = 0.0014) and TST, and for II genotype, between BMI and TST (p = 0.0071). II genotype was associated with severe obesity, while D allele carriers were associated with moderate undernutrition and moderate obesity. CONCLUSION BMI, MUAC, TST and serum protein levels are correlated with D allele carriers of ACE genes in children with moderate undernutrition and moderate obesity, whereas II genotype is an unfavorable prognostic factor corresponding to severe obesity and severe undernutrition.
Collapse
Affiliation(s)
| | - Claudia Bănescu
- Departement of Medical Genetics, University of Medicine and Pharmacy Tîrgu Mureş, Tîrgu Mureş, Romania.
| | - Carmen Duicu
- Pediatrics Clinic I, University of Medicine and Pharmacy Tîrgu Mureş, Tîrgu Mureş, Romania
| | - Septimiu Voidăzan
- Departement of Epidemiology, University of Medicine and Pharmacy Tîrgu Mureş, Tîrgu Mureş, Romania
| | - Claudiu Mărginean
- Departement of Obstetrics and Gynecology, University of Medicine and Pharmacy Tîrgu Mureş, Tîrgu Mureş, Romania
| |
Collapse
|
20
|
Cifani C, Micioni Di Bonaventura MV, Pucci M, Giusepponi ME, Romano A, Di Francesco A, Maccarrone M, D'Addario C. Regulation of hypothalamic neuropeptides gene expression in diet induced obesity resistant rats: possible targets for obesity prediction? Front Neurosci 2015; 9:187. [PMID: 26106286 PMCID: PMC4458694 DOI: 10.3389/fnins.2015.00187] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 05/11/2015] [Indexed: 12/11/2022] Open
Abstract
Several factors play a role in obesity (i.e., behavior, environment, and genetics) and epigenetic regulation of gene expression has emerged as a potential contributor in the susceptibility and development of obesity. To investigate the individual sensitivity to weight gain/resistance, we here studied gene transcription regulation of several hypothalamic neuropeptides involved in the control of energy balance in rats developing obesity (diet-induced obesity, DIO) or not (diet resistant, DR), when fed with a high fat diet. Rats have been followed up to 21 weeks of high fat diet exposure. After 5 weeks high fat diet exposure, the obese phenotype was developed and we observed a selective down-regulation of the orexigenic neuropeptide Y (NPY) and peroxisome proliferator-activated receptor gamma (PPAR-γ) genes. No changes were observed in the expression of the agouti-related protein (AgRP), as well as for all the anorexigenic genes under study. After long-term high fat diet exposure (21 weeks), NPY and PPAR-γ, as well as most of the genes under study, resulted not be different between DIO and DR, whereas a lower expression of the anorexigenic pro-opio-melanocortin (POMC) gene was observed in DIO rats when compared to DR rats. Moreover we observed that changes in NPY and POMC mRNA were inversely correlated with gene promoters DNA methylation. Our findings suggest that selective alterations in hypothalamic peptide genes regulation could contribute to the development of overweight in rats and that environmental factor, as in this animal model, might be partially responsible of these changes via epigenetic mechanism.
Collapse
Affiliation(s)
- Carlo Cifani
- Pharmacology Unit, School of Pharmacy, University of Camerino Camerino, Italy ; Intramural Research Program, National Institute on Drug Abuse/National Institutes of Health Baltimore, MD, USA
| | | | - Mariangela Pucci
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo Teramo, Italy
| | - Maria E Giusepponi
- Pharmacology Unit, School of Pharmacy, University of Camerino Camerino, Italy
| | - Adele Romano
- Department of Physiology and Pharmacology "V. Erspamer," Sapienza University of Rome Rome, Italy
| | - Andrea Di Francesco
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo Teramo, Italy
| | - Mauro Maccarrone
- Center of Integrated Research, Campus Bio-Medico University of Rome Rome, Italy ; European Center for Brain Research (CERC)/Santa Lucia Foundation Rome, Italy
| | - Claudio D'Addario
- Faculty of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo Teramo, Italy ; European Center for Brain Research (CERC)/Santa Lucia Foundation Rome, Italy
| |
Collapse
|
21
|
|
22
|
Pedrera-Zamorano JD, Roncero-Martin R, Lavado-Garcia JM, Calderon-Garcia JF, Rey-Sanchez P, Vera V, Martinez M, Moran JM. Segmental fat-free and fat mass measurements by bioelectrical impedance analysis in 2,224 healthy spanish women aged 18-85 years. Am J Hum Biol 2014; 27:468-74. [DOI: 10.1002/ajhb.22669] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2014] [Revised: 10/09/2014] [Accepted: 11/12/2014] [Indexed: 01/06/2023] Open
Affiliation(s)
- Juan D. Pedrera-Zamorano
- Metabolic Bone Diseases Research Group; Nursing Department, University of Extremadura; Caceres Spain
| | - Raul Roncero-Martin
- Metabolic Bone Diseases Research Group; Nursing Department, University of Extremadura; Caceres Spain
| | - Jesus M. Lavado-Garcia
- Metabolic Bone Diseases Research Group; Nursing Department, University of Extremadura; Caceres Spain
| | - Julian F. Calderon-Garcia
- Metabolic Bone Diseases Research Group; Nursing Department, University of Extremadura; Caceres Spain
| | - Purificacion Rey-Sanchez
- Metabolic Bone Diseases Research Group; Nursing Department, University of Extremadura; Caceres Spain
| | - Vicente Vera
- Metabolic Bone Diseases Research Group; Nursing Department, University of Extremadura; Caceres Spain
| | - Mariana Martinez
- Metabolic Bone Diseases Research Group; Nursing Department, University of Extremadura; Caceres Spain
| | - Jose M. Moran
- Metabolic Bone Diseases Research Group; Nursing Department, University of Extremadura; Caceres Spain
| |
Collapse
|
23
|
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarize recent advances in investigations of dietary factors, genetic factors, and their interactive effects on obesity and weight loss. RECENT FINDINGS Even with a tremendous body of research conducted, controversy still abounds regarding the relative effectiveness of various weight-loss diets. Recent advances in genome-wide association studies have made great strides in unraveling the genetic basis of regulation of body weight. In prospective cohorts, reproducible evidence is emerging to show interactions between genetic factors and dietary factors such as sugar-sweetened beverage on obesity. In randomized clinical trials, individuals' genotypes have also been found to modify diet interventions on weight loss, weight maintenance, and changes in related metabolic traits such as lipids, insulin resistance, and blood pressure. However, replication, functional exploration, and translation of the findings into personalized diet interventions remain the chief challenges. SUMMARY Preliminary but promising data have emerged to lend support to gene-diet interaction in determining weight loss and maintenance; and studies in the area hold great promise to inform future personalized diet interventions on the reduction of obesity and related health problems.
Collapse
Affiliation(s)
- Lu Qi
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
24
|
Dietary Management and Genetic Predisposition. Curr Nutr Rep 2013. [DOI: 10.1007/s13668-013-0050-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
25
|
Garver WS, Newman SB, Gonzales-Pacheco DM, Castillo JJ, Jelinek D, Heidenreich RA, Orlando RA. The genetics of childhood obesity and interaction with dietary macronutrients. GENES AND NUTRITION 2013; 8:271-87. [PMID: 23471855 DOI: 10.1007/s12263-013-0339-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Accepted: 02/22/2013] [Indexed: 12/21/2022]
Abstract
The genes contributing to childhood obesity are categorized into three different types based on distinct genetic and phenotypic characteristics. These types of childhood obesity are represented by rare monogenic forms of syndromic or non-syndromic childhood obesity, and common polygenic childhood obesity. In some cases, genetic susceptibility to these forms of childhood obesity may result from different variations of the same gene. Although the prevalence for rare monogenic forms of childhood obesity has not increased in recent times, the prevalence of common childhood obesity has increased in the United States and developing countries throughout the world during the past few decades. A number of recent genome-wide association studies and mouse model studies have established the identification of susceptibility genes contributing to common childhood obesity. Accumulating evidence suggests that this type of childhood obesity represents a complex metabolic disease resulting from an interaction with environmental factors, including dietary macronutrients. The objective of this article is to provide a review on the origins, mechanisms, and health consequences of obesity susceptibility genes and interaction with dietary macronutrients that predispose to childhood obesity. It is proposed that increased knowledge of these obesity susceptibility genes and interaction with dietary macronutrients will provide valuable insight for individual, family, and community preventative lifestyle intervention, and eventually targeted nutritional and medicinal therapies.
Collapse
Affiliation(s)
- William S Garver
- Department of Biochemistry and Molecular Biology, School of Medicine, University of New Mexico Health Sciences Center, Albuquerque, NM, 87131-0001, USA,
| | | | | | | | | | | | | |
Collapse
|
26
|
Leamy LJ, Kelly SA, Hua K, Pomp D. Exercise and diet affect quantitative trait loci for body weight and composition traits in an advanced intercross population of mice. Physiol Genomics 2012; 44:1141-53. [PMID: 23048196 PMCID: PMC3544482 DOI: 10.1152/physiolgenomics.00115.2012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 10/04/2012] [Indexed: 12/25/2022] Open
Abstract
Driven by the recent obesity epidemic, interest in understanding the complex genetic and environmental basis of body weight and composition is great. We investigated this by searching for quantitative trait loci (QTLs) affecting a number of weight and adiposity traits in a G(10) advanced intercross population produced from crosses of mice in inbred strain C57BL/6J with those in a strain selected for high voluntary wheel running. The mice in this population were fed either a high-fat or a control diet throughout the study and also measured for four exercise traits prior to death, allowing us to test for pre- and postexercise QTLs as well as QTL-by-diet and QTL-by-exercise interactions. Our genome scan uncovered a number of QTLs, of which 40% replicated QTLs previously found for similar traits in an earlier (G(4)) generation. For those replicated QTLs, the confidence intervals were reduced from an average of 19 Mb in the G(4) to 8 Mb in the G(10). Four QTLs on chromosomes 3, 8, 13, and 18 were especially prominent in affecting the percentage of fat in the mice. About of all QTLs showed interactions with diet, exercise, or both, their genotypic effects on the traits showing a variety of patterns depending on the diet or level of exercise. It was concluded that the indirect effects of these QTLs provide an underlying genetic basis for the considerable variability in weight or fat loss typically found among individuals on the same diet and/or exercise regimen.
Collapse
Affiliation(s)
- Larry J Leamy
- Department of Biology, University of North Carolina at Charlotte, Charlotte, North Carolina 28223, USA.
| | | | | | | |
Collapse
|
27
|
Vasilopoulos Y, Sarafidou T, Kotsa K, Papadimitriou M, Goutzelas Y, Stamatis C, Bagiatis V, Tsekmekidou X, Yovos JG, Mamuris Z. VDR TaqI is associated with obesity in the Greek population. Gene 2012; 512:237-9. [PMID: 23103831 DOI: 10.1016/j.gene.2012.10.044] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Revised: 10/01/2012] [Accepted: 10/11/2012] [Indexed: 01/22/2023]
Abstract
The prevalence of obesity has increased dramatically during the last thirty years in western countries with severe complications for health and economy. Obesity is the outcome of the strong interplay between genetic and environmental factors and is therefore widely expected that the discovery of the many genetic factors underlying the heritable risk of obesity will contribute critically to our basic knowledge of the disease etiopathogenesis and the identification of new targets for therapeutic intervention. The aim of the present study was to assess the genetic contribution of known polymorphisms in two genes that are linked to the pathogenetic mechanism of obesity. Analysis of vitamin D receptor (VDR) TaqI (rs731236; T/C) and fat mass and obesity-associated (FTO) (rs9939609; A/T) [corrected] polymorphisms in 82 obesity subjects and 102 controls showed significant association for VDR TaqI 'T' allele and obesity (OR: 2.07; 1.123-3.816; P=0.019), contributing to an elevated BMI of 3kg/m(2) per risk allele. No association was observed for the FTO polymorphism. These results further support a role for VDR as risk factor for obesity and suggest its further validation in larger independent populations as well as highlight a target for functional analysis towards therapeutic intervention in obese individuals.
Collapse
Affiliation(s)
- Yiannis Vasilopoulos
- Department of Biochemistry and Biotechnology, University of Thessaly, Larissa, Greece.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Qi L. Gene-Diet Interactions in Complex Disease: Current Findings and Relevance for Public Health. Curr Nutr Rep 2012; 1:222-227. [PMID: 23139897 DOI: 10.1007/s13668-012-0029-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Rates of obesity and related complex diseases such as type 2 diabetes and cardiovascular disease have climbed sharply over the past decades, in parallel with shift from principally more active lifestyle and nutritionally dense tradition diet to sedentary lifestyle and more energy-dense, Western-pattern diet. In the past few years, advances in genotyping technology and in particular a number of large-scale genome-wide association studies have made great strides in unraveling the genetic basis of complex diseases; and the growing inventory of genetic variation is facilitating efforts to investigate gene-diet interactions. Understanding gene-diet interaction has the potential to promote diet modifications on the basis of genetic makeup. Several recent large-scale studies found reproducible evidence showing consumption of sugar sweetened beverages or dietary patterns might modulate genetic predisposition to obesity or cardiovascular disease. Analyses in randomized trials also showed that genetic markers for obesity, diabetes, or cardiovascular disease might modify the metabolic response to weight-loss diets. However, little of the knowledge about gene-diet interaction has been applied in public health practice; and opinion on how genetic testing services are offered and interpreted is still divided. This review will summarize recent findings regarding obesogenic diet, genetic susceptibility, and gene-diet interactions for obesity and related complex disorders and will discuss the potential impact of these findings on public health practice.
Collapse
Affiliation(s)
- Lu Qi
- Department of Nutrition, Harvard School of Public Health, Boston, Massachusetts ; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
29
|
Increased hypothalamic inflammation associated with the susceptibility to obesity in rats exposed to high-fat diet. EXPERIMENTAL DIABETES RESEARCH 2012; 2012:847246. [PMID: 22844271 PMCID: PMC3401545 DOI: 10.1155/2012/847246] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Accepted: 05/14/2012] [Indexed: 01/11/2023]
Abstract
Inflammation has been implicated in the hypothalamic leptin and insulin resistance resulting defective food intake during high fat diet period. To investigate hypothalamic inflammation in dietary induced obesity (DIO) and obesity resistant (DIO-R) rats, we established rat models of DIO and DIO-R by feeding high fat diet for 10 weeks. Then we switched half of DIO and DIO-R rats to chow food and the other half to high fat diet for the following 8 weeks to explore hypothalamic inflammation response to the low fat diet intervention. Body weight, caloric intake, HOMA-IR, as well as the mRNA expression of hypothalamic TLR4, NF-κB, TNF-α, IL-1β, and IL-6 in DIO/HF rats were significantly increased compared to DIO-R/HF and CF rats, whereas IL-10 mRNA expression was lower in both DIO/HF and DIO-R/HF rats compared with CF rats. Switching to chow food from high fat diet reduced the body weight and improved insulin sensitivity but not affecting the expressions of studied inflammatory genes in DIO rats. Take together, upregulated hypothalamic inflammation may contribute to the overeating and development of obesity susceptibility induced by high fat diet. Switching to chow food had limited role in correcting hypothalamic inflammation in DIO rats during the intervention period.
Collapse
|
30
|
Abstract
The prevalence of the metabolic syndrome (MetS) has increased rapidly in North America in recent years. Presently, the MetS is found in 34.3% of the population, and the prevalence is likely to continue to increase in parallel with the obesity epidemic. Losing weight and long-term maintenance of the weight loss are primary targets to beneficially reverse all of the components of the MetS. This paper reviews the clinical and experimental evidence for the reversal of the metabolic complications related to the MetS that follows a sustained weight loss.
Collapse
Affiliation(s)
- Annie Ferland
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Campus, Anschutz Medical Center, 12801 E. 17th Avenue, Aurora, CO 80045, USA
| | | |
Collapse
|
31
|
Zhang J, Himes JH, Hannan PJ, Arcan C, Smyth M, Rock BH, Story M. Summer effects on body mass index (BMI) gain and growth patterns of American Indian children from kindergarten to first grade: a prospective study. BMC Public Health 2011; 11:951. [PMID: 22192795 PMCID: PMC3260204 DOI: 10.1186/1471-2458-11-951] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 12/23/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Overweight and obesity are highly prevalent among American Indian children, especially those living on reservations. There is little scientific evidence about the effects of summer vacation on obesity development in children. The purpose of this study was to investigate the effects of summer vacation between kindergarten and first grade on growth in height, weight, and body mass index (BMI) for a sample of American Indian children. METHODS Children had their height and weight measured in four rounds of data collection (yielded three intervals: kindergarten, summer vacation, and first grade) as part of a school-based obesity prevention trial (Bright Start) in a Northern Plains Indian Reservation. Demographic variables were collected at baseline from parent surveys. Growth velocities (Z-score units/year) for BMI, weight, and height were estimated and compared for each interval using generalized linear mixed models. RESULTS The children were taller and heavier than median of same age counterparts. Height Z-scores were positively associated with increasing weight status category. The mean weight velocity during summer was significantly less than during the school year. More rapid growth velocity in height during summer than during school year was observed. Obese children gained less adjusted-BMI in the first grade after gaining more than their counterparts during the previous two intervals. No statistically significant interval effects were found for height and BMI velocities. CONCLUSIONS There was no indication of a significant summer effect on children's BMI. Rather than seasonal or school-related patterns, the predominant pattern indicated by weight-Z and BMI-Z velocities might be related to age or maturation. TRIAL REGISTRATION Bright Start: Obesity Prevention in American Indian Children Clinical Trial Govt ID# NCT00123032.
Collapse
Affiliation(s)
- Jianduan Zhang
- Division of Epidemiology and Community Health, School of, Public Health, University of Minnesota, 1300 South 2nd Street, Suite 300, Minneapolis, MN 55454, USA
- Department of Maternal and Children Care and Adolescent Health, School of Public Health, Huazhong University of Science and Technology, Wuhan, Hubei, China 430030
| | - John H Himes
- Division of Epidemiology and Community Health, School of, Public Health, University of Minnesota, 1300 South 2nd Street, Suite 300, Minneapolis, MN 55454, USA
| | - Peter J Hannan
- Division of Epidemiology and Community Health, School of, Public Health, University of Minnesota, 1300 South 2nd Street, Suite 300, Minneapolis, MN 55454, USA
| | - Chrisa Arcan
- Division of Epidemiology and Community Health, School of, Public Health, University of Minnesota, 1300 South 2nd Street, Suite 300, Minneapolis, MN 55454, USA
| | - Mary Smyth
- Division of Epidemiology and Community Health, School of, Public Health, University of Minnesota, 1300 South 2nd Street, Suite 300, Minneapolis, MN 55454, USA
| | - Bonnie Holy Rock
- Division of Epidemiology and Community Health, School of, Public Health, University of Minnesota, 1300 South 2nd Street, Suite 300, Minneapolis, MN 55454, USA
| | - Mary Story
- Division of Epidemiology and Community Health, School of, Public Health, University of Minnesota, 1300 South 2nd Street, Suite 300, Minneapolis, MN 55454, USA
| |
Collapse
|
32
|
Garver WS. Gene-diet interactions in childhood obesity. Curr Genomics 2011; 12:180-9. [PMID: 22043166 PMCID: PMC3137003 DOI: 10.2174/138920211795677903] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Revised: 02/17/2011] [Accepted: 02/28/2011] [Indexed: 12/31/2022] Open
Abstract
Childhood overweight and obesity have reached epidemic proportions worldwide, and the increase in weight-associated co-morbidities including premature type 2 diabetes mellitus (T2DM) and atherosclerotic cardiovascular disease will soon become major healthcare and economic problems. A number of studies now indicate that the childhood obesity epidemic which has emerged during the past 30 years is a complex multi-factorial disease resulting from interaction of susceptibility genes with an obesogenic environment. This review will focus on gene-diet interactions suspected of having a prominent role in promoting childhood obesity. In particular, the specific genes that will be presented (FTO, MC4R, and NPC1) have recently been associated with childhood obesity through a genome-wide association study (GWAS) and were shown to interact with nutritional components to increase weight gain. Although a fourth gene (APOA2) has not yet been associated with childhood obesity, this review will also present information on what now represents the best characterized gene-diet interaction in promoting weight gain.
Collapse
Affiliation(s)
- William S Garver
- Department of Biochemistry and Molecular Biology, The University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| |
Collapse
|
33
|
Nascimento VG, Schoeps DDO, Souza SBD, Souza JMPD, Leone C. Risco de sobrepeso e excesso de peso em crianças de pré-escolas privadas e filantrópicas. Rev Assoc Med Bras (1992) 2011; 57:657-61. [DOI: 10.1590/s0104-42302011000600012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2011] [Accepted: 09/06/2011] [Indexed: 11/22/2022] Open
|
34
|
Risk of overweight and obesity in preschoolers attending private and philanthropic schools. Rev Assoc Med Bras (1992) 2011. [DOI: 10.1016/s0104-4230(11)70130-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
35
|
González-Castejón M, Rodriguez-Casado A. Dietary phytochemicals and their potential effects on obesity: A review. Pharmacol Res 2011; 64:438-55. [DOI: 10.1016/j.phrs.2011.07.004] [Citation(s) in RCA: 207] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Accepted: 07/11/2011] [Indexed: 12/20/2022]
|
36
|
Costa MC, Brito LL, Barbosa PJB, Lessa I. Adiponectina e baixo risco cardiometabólico em obesas. ACTA ACUST UNITED AC 2011; 55:146-54. [DOI: 10.1590/s0004-27302011000200007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Accepted: 02/16/2011] [Indexed: 11/22/2022]
Abstract
OBJETIVO: Investigar a associação entre níveis elevados de adiponectina plasmática (AdipoQ) e baixo risco cardiometabólico (RCM) em obesas (MOb). SUJEITOS E MÉTODOS: Estudo caso-controle, pareado pela idade com 306 MOb, IMC > 30 kg/m², sendo 66 (21,6%) casos - todos não hipertensos, normolipídicos e não diabéticos - realizados em ambulatórios referência do Sistema Único de Saúde (SUS) para obesidade em Salvador. Dados secundários foram coleta-dos dos prontuários médicos, primários e complementares em laboratório de patologia clínica. Valores de AdipoQ > 10,00 µg/mL (> 3º quartil) foram considerados elevados. Foram realizadas análises: descritiva, bivariada e regressão logística condicional. RESULTADOS: Associação positiva, estatisticamente significante (ES), entre baixo RCM e AdipoQ > 10,00 µg/mL em MOb ativas fisicamente (OR= 5,1; IC95%: 1,8-14,3), não fumantes (OR= 3,6; IC95%: 1,6-8,4). CONCLUSÃO: Este estudo sugere que MOb ativas fisicamente, não fumantes e com níveis mais elevados de AdipoQ apresentam mais chances de baixo RCM. Torna-se importante reforçar, por meio de políticas públicas, o estímulo à adoção de comportamentos saudáveis.
Collapse
|
37
|
Tapsell L, Batterham M, Huang XF, Tan SY, Teuss G, Charlton K, Oshea J, Warensjö E. Short term effects of energy restriction and dietary fat sub-type on weight loss and disease risk factors. Nutr Metab Cardiovasc Dis 2010; 20:317-325. [PMID: 19570664 DOI: 10.1016/j.numecd.2009.04.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2008] [Revised: 03/31/2009] [Accepted: 04/06/2009] [Indexed: 12/31/2022]
Abstract
BACKGROUND AND AIMS Decreasing energy intake relative to energy expenditure is the indisputable tenet of weight loss. In addition to caloric restriction modification of the type of dietary fat may provide further benefits. The aim of the present study was to examine the effect of energy restriction alone and with dietary fat modification on weight loss and adiposity, as well as on risk factors for obesity related disease. METHODS AND RESULTS One-hundred and fifty overweight men and women were randomized into a 3month controlled trial with four low fat (30% energy) dietary arms: (1) isocaloric (LF); (2) isocaloric with 10% polyunsaturated fatty acids (LF-PUFA); (3) low calorie (LF-LC) (-2MJ); (4) low calorie with 10% PUFA (LF-PUFA-LC). Primary outcomes were changes in body weight and body fat and secondary outcomes were changes in fasting levels of leptin, insulin, glucose, lipids and erythrocyte fatty acids. Changes in dietary intake were assessed using 3day food records. One-hundred and twenty-two participants entered the study and 95 completed the study. All groups lost weight and body fat (P<0.0001 time effect for both), but the LC groups lost more weight (P=0.026 for diet effect). All groups reduced total cholesterol levels (P<0.0001 time effect and P=0.017 intervention effect), but the LC and PUFA groups were better at reducing triacylglycerol levels (P=0.056 diet effect). HDL increased with LF-LC and LF-PUFA but not with LF-PUFA-LC (0.042 diet effect). The LF and LF-LC groups reported greater dietary fat reductions than the two PUFA groups (P=0.043). CONCLUSION Energy restriction has the most potent effect on weight loss and lipids, but fat modification is also beneficial when energy restriction is more modest.
Collapse
Affiliation(s)
- L Tapsell
- Smart Foods Centre, University of Wollongong, Wollongong, NSW 2522, Australia
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Lazarou C, Kouta C. The role of nurses in the prevention and management of obesity. ACTA ACUST UNITED AC 2010; 19:641-7. [DOI: 10.12968/bjon.2010.19.10.48203] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Chrystalleni Lazarou
- Harokopio University, Department, Nutrition and Dietetics, Athens, Greece; CyprusUniversity of Technology, Department of Agricultural Sciences, Biotechnology & Food Science, Cyprus
| | - Christiana Kouta
- Cyprus University of Technology, Department of Nursing, School of Health Sciences, Cyprus
| |
Collapse
|
39
|
Joffe YT, van der Merwe L, Carstens M, Collins M, Jennings C, Levitt NS, Lambert EV, Goedecke JH. Tumor necrosis factor-alpha gene -308 G/A polymorphism modulates the relationship between dietary fat intake, serum lipids, and obesity risk in black South African women. J Nutr 2010; 140:901-7. [PMID: 20335630 DOI: 10.3945/jn.109.109355] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The prevalence of obesity and related disease risk is high in black South African (SA) women, possibly influenced by the dietary transition associated with urbanization. This study explored interactions between dietary fat intake and the tumor necrosis factor-alpha (TNFA) -308 G/A polymorphism on obesity, insulin resistance, and serum lipid concentrations in urbanized black SA women. Normal-weight (n = 105) and obese (n = 118) women underwent measurements of body composition, fat distribution, fasting serum lipids, glucose and insulin concentrations, and dietary intake. Participants were genotyped for the functional TNFA -308 G/A polymorphism. The genotype or allele frequency of the TNFA -308 G/A polymorphism did not differ between the BMI groups. However, when dietary fat intake was 30% of total energy intake [percentage energy (%E)], the odds of being obese with the TNFA GA+AA genotype was only 12% of that with GG, but increasing intake of dietary fat (%E) was associated with a significantly faster rate of increase in obesity risk in women with the TNFA GA+AA genotype compared with those with the GG genotype (P = 0.036). There were significant diet-gene interactions between alpha-linolenic acid (%E) and the total cholesterol:HDL-cholesterol ratio (P = 0.036), and PUFA (%E) and LDL cholesterol levels (P = 0.026), with participants with the A allele being more responsive to changes in relative fat intake. The TNFA -308 G/A polymorphism modified the relationship between dietary fat intake, obesity risk, and serum lipid concentrations in black SA women.
Collapse
Affiliation(s)
- Yael T Joffe
- University of Cape Town/Medical Research Council Research Unit for Exercise Science and Sports Medicine,, Cape Town, South Africa
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Balaban G, Motta MEFA, Silva GAP. Early weaning and other potential risk factors for overweight among preschool children. Clinics (Sao Paulo) 2010; 65:181-7. [PMID: 20186302 PMCID: PMC2827705 DOI: 10.1590/s1807-59322010000200010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2009] [Accepted: 11/26/2009] [Indexed: 11/21/2022] Open
Abstract
OBJECTIVE To investigate whether early weaning constitutes a risk factor for overweight at preschool age and to identify other factors that affect this association. METHODS This was a case-control study of 366 children aged 2 to 6 years (176 boys and 190 girls) from three cities. The case group comprised overweight children, as defined by body mass index (BMI) for age greater than or equal to the 85(th) percentile. The main exposure analyzed was early weaning (exclusive or predominant breastfeeding for less than four months). RESULTS Early weaning was a significant risk factor for overweight in univariate analysis (OR = 1.69; 95% CI: 1.10-2.60; p = 0.02), but not in multivariate analysis (OR = 1.42; 95% CI: 0.86-2.34; p = 0.17). Maternal overweight, birth weight > or = 3,500 g and sedentarism were the main risk factors for overweight in multivariate analysis. DISCUSSION In our study, the protective effect of breastfeeding against overweight was only shown in univariate analysis; it did not persist after controlling for other variables. It is possible that breastfeeding has only a small protective role against overweight in comparison with other variables of greater importance. CONCLUSION Our results suggest that the potential protective effect of breastfeeding against overweight among preschool children is weaker than genetic and other environmental factors.
Collapse
Affiliation(s)
- Geni Balaban
- Federal University of Ceará and School of Medicine of Juazeiro do Norte - Ceará/PE, Brazil.
| | | | | |
Collapse
|
41
|
Abstract
Stigma and discrimination toward obese persons are pervasive and pose numerous consequences for their psychological and physical health. Despite decades of science documenting weight stigma, its public health implications are widely ignored. Instead, obese persons are blamed for their weight, with common perceptions that weight stigmatization is justifiable and may motivate individuals to adopt healthier behaviors. We examine evidence to address these assumptions and discuss their public health implications. On the basis of current findings, we propose that weight stigma is not a beneficial public health tool for reducing obesity. Rather, stigmatization of obese individuals threatens health, generates health disparities, and interferes with effective obesity intervention efforts. These findings highlight weight stigma as both a social justice issue and a priority for public health.
Collapse
Affiliation(s)
- Rebecca M Puhl
- Director of Research and Weight Stigma Initiatives, Rudd Center for Food Policy and Obesity, Yale University, 309 Edwards St, New Haven, CT 06520-8369, USA.
| | | |
Collapse
|
42
|
Affiliation(s)
- Dolores Corella
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
- Genetic and Molecular Epidemiology Unit and CIBER Fisiopatología de la Obesidad y Nutrición as an initiative of the ISCIII. School of Medicine. University of Valencia, Valencia, Spain
| | - Jose M. Ordovas
- Nutrition and Genomics Laboratory, JM-USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA, USA
- Department of Cardiovascular Epidemiology and Population Genetics, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| |
Collapse
|
43
|
Childhood overweight and obesity among Kenyan pre-school children: association with maternal and early child nutritional factors. Public Health Nutr 2009; 13:496-503. [DOI: 10.1017/s136898000999187x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
AbstractObjectiveTo report on the prevalence of overweight and obesity among pre-school children in Kenya and examine the associations between childhood overweight and selected maternal and child-related factors.DesignDemographic Health Survey data, multistage stratified cluster sampling methodology.SettingRural and urban areas of Kenya.SubjectsA total of 1495 children between the ages of 3 and 5 years in Kenya.ResultsOver 30 % of the children were stunted, approximately 16 % were underweight, 4 % were wasted, approximately 18 % were overweight and 4 % were obese; 8 % were both overweight/obese and stunted. Maternal overweight and obesity, higher levels of maternal education, being a large or very large child at birth, and being stunted were each associated with higher odds of overweight and obesity among Kenyan children. Older children and large household size were each associated with lower odds of overweight and obesity among Kenyan children.ConclusionsThe analysis demonstrates the presence of under- and overnutrition among Kenyan pre-school children and the importance of focusing on expanding efforts to prevent and treat malnutrition within this population. It also identifies some of the modifiable factors that can be targeted in these efforts.
Collapse
|
44
|
Morris MJ. Early life influences on obesity risk: maternal overnutrition and programming of obesity. Expert Rev Endocrinol Metab 2009; 4:625-637. [PMID: 30780787 DOI: 10.1586/eem.09.45] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
While adult lifestyle factors undoubtedly contribute to the incidence of obesity and its attendant disorders, mounting evidence suggests that programming of obesity may occur following over-nutrition during development. As hypothalamic control of appetite and energy expenditure is set early in life and can be perturbed by certain exposures, such as undernutrition and altered metabolic and hormonal signals, in utero exposure to maternal obesity-related changes may contribute to programming of obesity in offspring. Data from animal studies indicate both intrauterine and postnatal environments are critical determinants of the development of pathways regulating energy homeostasis. This review summarizes recent evidence of the impact of maternal obesity on subsequent obesity risk, paying particular attention to the hypothalamic regulation of appetite and markers of metabolic control. The extraordinary rise in the rates of maternal obesity underlines an urgent need to investigate the mechanisms contributing to its transgenerational effects.
Collapse
Affiliation(s)
- Margaret J Morris
- a Department of Pharmacology, School of Medical Sciences, University of New South Wales, NSW 2052, Australia.
| |
Collapse
|
45
|
Changes in white muscle transcriptome induced by dietary energy levels in two lines of rainbow trout (Oncorhynchus mykiss) selected for muscle fat content. Br J Nutr 2009; 103:629-42. [PMID: 19874644 DOI: 10.1017/s0007114509992340] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Energy intake and genetic background are major determinants of muscle fat content in most animals, including man. We combined genetic selection and dietary energy supply to study the metabolic pathways involved in genetic and nutritional control of fat deposition in the muscle of rainbow trout (Oncorhynchus mykiss). Two experimental lines of rainbow trout, selected for lean (L) or fat (F) muscle, were fed with diets containing either 10 or 23 % lipids from the first feeding, up to 6 months. At the end of the trial, trout exhibited very different values of muscle fat content (from 4.2 to 10.1 % wet weight). Using microarrays made from a rainbow trout multi-tissue cDNA library, we analysed the molecular changes occurring in the muscle of the two lines when fed the low-energy or high-energy diet. The results from microarray analysis revealed that eleven metabolism-related genes were differentially expressed according to the diet while selection resulted in expression change for twenty-six genes. The most striking observation was the increased level of transcripts encoding the VLDL receptor and fatty acid translocase/CD36 following both the high-fat diet and upward selection for muscle fat content, suggesting that these two genes are relevant molecular markers of fat deposition in the white muscle of rainbow trout.
Collapse
|
46
|
van Velzen EJJ, Westerhuis JA, van Duynhoven JPM, van Dorsten FA, Grün CH, Jacobs DM, Duchateau GSMJE, Vis DJ, Smilde AK. Phenotyping tea consumers by nutrikinetic analysis of polyphenolic end-metabolites. J Proteome Res 2009; 8:3317-30. [PMID: 19374449 DOI: 10.1021/pr801071p] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
An integration of metabolomics and pharmacokinetics (or nutrikinetics) is introduced as a concept to describe a human study population with different metabolic phenotypes following a nutritional intervention. The approach facilitates an unbiased analysis of the time-response of body fluid metabolites from crossover designed intervention trials without prior knowledge of the underlying metabolic pathways. The method is explained for the case of a human intervention study in which the nutrikinetic analysis of polyphenol-rich black tea consumption was performed in urine over a period of 48 h. First, multilevel PLS-DA analysis was applied to the urinary 1H NMR profiles to select the most differentiating biomarkers between the verum and placebo samples. Then, a one-compartment nutrikinetic model with first-order excretion, a lag time, and a baseline function was fitted to the time courses of these selected biomarkers. The nutrikinetic model used here fully exploits the crossover structure in the data by fitting the data from both the treatment period and the placebo period simultaneously. To demonstrate the procedure, a selected set of urinary biomarkers was used in the model fitting. These metabolites include hippuric acid, 4-hydroxyhippuric acid and 1,3-dihydroxyphenyl-2-O-sulfate and derived from microbial fermentation of polyphenols in the gut. Variations in urinary excretion between- and within the subjects were observed, and used to provide a phenotypic description of the test population.
Collapse
Affiliation(s)
- Ewoud J J van Velzen
- Biosystems Data Analysis, Swammerdam Institute for Life Sciences, Universiteit van Amsterdam, Nieuwe Achtergracht 166, 1018 WV Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
|
48
|
Coxon GD, Furman BL, Harvey AL, McTavish J, Mooney MH, Arastoo M, Kennedy AR, Tettey JM, Waigh RD. Benzylguanidines and Other Galegine Analogues Inducing Weight Loss in Mice. J Med Chem 2009; 52:3457-63. [DOI: 10.1021/jm8011933] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Geoffrey D. Coxon
- Strathclyde Institute for Pharmacy and Biomedical Sciences and Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, United Kingdom
| | - Brian L. Furman
- Strathclyde Institute for Pharmacy and Biomedical Sciences and Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, United Kingdom
| | - Alan L. Harvey
- Strathclyde Institute for Pharmacy and Biomedical Sciences and Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, United Kingdom
| | - John McTavish
- Strathclyde Institute for Pharmacy and Biomedical Sciences and Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, United Kingdom
| | - Mark H. Mooney
- Strathclyde Institute for Pharmacy and Biomedical Sciences and Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, United Kingdom
| | - Mahmoud Arastoo
- Strathclyde Institute for Pharmacy and Biomedical Sciences and Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, United Kingdom
| | - Alan R. Kennedy
- Strathclyde Institute for Pharmacy and Biomedical Sciences and Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, United Kingdom
| | - Justice M. Tettey
- Strathclyde Institute for Pharmacy and Biomedical Sciences and Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, United Kingdom
| | - Roger D. Waigh
- Strathclyde Institute for Pharmacy and Biomedical Sciences and Department of Pure and Applied Chemistry, University of Strathclyde, Glasgow, United Kingdom
| |
Collapse
|
49
|
Newby PK. Plant foods and plant-based diets: protective against childhood obesity? Am J Clin Nutr 2009; 89:1572S-1587S. [PMID: 19321559 DOI: 10.3945/ajcn.2009.26736g] [Citation(s) in RCA: 104] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The objective of this article is to review the epidemiologic literature examining the role of plant foods and plant-based diets in the prevention of childhood obesity. Available data suggest a protective effect of ready-to-eat cereal on risk of obesity, although prospective studies are still needed. Studies on fruit and vegetables; grains other than cereal; high-protein foods, including beans, legumes, and soy; fiber; and plant-based dietary patterns are inconsistent or generally null. The evidence base is limited, and most studies are fraught with methodologic limitations, including cross-sectional design, inadequate adjustment for potential confounders, and lack of consideration of reporting errors, stage of growth, and genetic influences. Well-designed prospective studies are needed. The lack of evidence showing an association between plant-based diets and childhood obesity does not mean that such diets should not be encouraged. Plant foods are highlighted in the Dietary Guidelines for Americans, and children do not meet the current recommendations for most plant foods. Although the advice to consume a plant-based, low-energy-dense diet is sound, ethical questions arise concerning the relatively high price of these diets in the United States and the way in which such diets are perceived in other parts of the world. Reducing the burden of childhood obesity, eliminating health disparities, and preventing the further spread of the disease around the globe will require not only policy interventions to ensure that plant foods are affordable and accessible to children of all income levels but also awareness of sociocultural norms that affect consumption.
Collapse
Affiliation(s)
- P K Newby
- Boston University School of Medicine, Boston University School of Public Health, Boston, MA, USA.
| |
Collapse
|
50
|
Interaction effects between total energy and macronutrient intakes and angiotensin-converting enzyme 1 (ACE) I/D polymorphism on adiposity-related phenotypes in toddlers and preschoolers: the Growth, Exercise and Nutrition Epidemiological Study in preSchoolers (GENESIS). Br J Nutr 2008; 100:1333-40. [DOI: 10.1017/s0007114508988759] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The aim of the present study was to investigate the interaction between the angiotensin-converting enzyme 1 (ACE) I/D polymorphism and energy and macronutrient intakes on adiposity-related phenotypes among toddlers and preschoolers. A representative sample of 2374 Greek children aged 1 to 5 years old was examined (Growth, Exercise and Nutrition Epidemiological Study in preSchoolers (GENESIS)). Dietary and anthropometric (i.e. BMI, waist circumference (WC)) assessments were carried out using standard procedures. DNA samples were obtained from 2102 children and were genotyped for theACEI/D polymorphism. Among the entire population, 17 % were ‘at risk of overweight’ and a similar percentage were ‘overweight’. The frequencies of the II, ID and DD genotypes were 16, 46 and 38 %, respectively. Significant interactions were found between theACEI/D polymorphism and total energy intake on WC (P = 0·004 for interaction) and theACEI/D polymorphism and protein intake on BMI and being overweight (P < 0·05 for interaction). Furthermore, it was found that theACEI/D polymorphism may modify the effect of fat intake on WC and BMI, but this interaction disappeared after adjustment for additional potential confounders. Stratified analyses revealed that total energy is correlated with WC and protein intake is associated with BMI and being overweight only among carriers of the D-allele (i.e. DD or ID genotypes). These results suggest that theACEI/D polymorphism may act as a modifying factor in the response of adiposity-related phenotypes to diet.
Collapse
|