1
|
Bray GA, Qi L, Sacks FM. Is There an Ideal Diet? Some Insights from the POUNDS Lost Study. Nutrients 2024; 16:2358. [PMID: 39064800 PMCID: PMC11280300 DOI: 10.3390/nu16142358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 07/09/2024] [Accepted: 07/17/2024] [Indexed: 07/28/2024] Open
Abstract
Diets for weight loss have a long history but an ideal one has not yet been clearly identified. To compare low-fat and lower carbohydrate diets, we designed The Preventing Overweight by Novel Dietary Strategies (POUNDS) Lost study. This is a 2 × 2 factorial study with diets of 20% or 40% fat and 15% or 25% protein with a graded carbohydrate intake of 35, 45, 55 and 65%. Weight loss, overall, was modest at nearly 6% with all four diets, and no significant dietary difference. The variability in weight loss in each diet group was significant, ranging from greater than 20% to a small weight gain. Studies of genetic variations in relation to weight loss showed that the diet that was selected could significantly affect weight loss, emphasizing that there is no ideal diet and more than one diet can be used to treat obesity. Weight loss was also influenced by the level of baseline triiodothyronine or thyroxine, and baseline carbohydrate and insulin resistance. Achieving a stable Health Eating Food Diversity Index, eating more protein, eating more fiber, engaging in more physical activity, sleeping better and eating less ultra-processed foods were beneficial strategies for weight loss in this trial. Although there is no "ideal diet", both the DASH diet and the Mediterranean diet have clinical trials showing their significant benefit for cardiovascular risk factors. Finally, the lesson of the "Last Chance Diet", which recommended a diet with protein from gelatin, proved that some diets could be hazardous.
Collapse
Affiliation(s)
- George A. Bray
- Department of Clinical Obesity, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, USA
| | - Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orlean, LA 70112, USA;
| | - Frank M. Sacks
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA;
| |
Collapse
|
2
|
Bennett KA, Sutherland C, Savage AL. A systematic review and meta-analysis of environmental contaminant exposure impacts on weight loss and glucose regulation during calorie-restricted diets in preclinical studies: Persistent organic pollutants may impede glycemic control. Biochem Pharmacol 2024; 225:116300. [PMID: 38782075 DOI: 10.1016/j.bcp.2024.116300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024]
Abstract
Epidemiological evidence links chemical exposure with type 2 diabetes (T2DM) risk and prevalence. Chemical exposure may therefore also limit success of weight loss or restoration of glycemic control during calorie restricted diets. Few human studies examine this hypothesis. This systematic review and clustered meta-analysis examines preclinical evidence that exposure to anthropogenic environmental contaminants impedes weight loss and resumption of glycemic control during calorie restriction. Of five eligible papers from 212 unique citations, four used C57BL/6 mice and one used Sprague Dawley rats. In four the animals received high fat diets to induce obesity and impaired glycemic control. All examined persistent organic pollutants (POPs). Polychlorinated biphenyl (PCB) 77 exposure did not affect final mass (standardised mean difference (SMD) = -0.35 [-1.09, 0.39]; n = 5 (experiments); n = 3 (papers)), or response to insulin in insulin tolerance tests (SMD = -1.54 [-3.25, 0.16] n = 3 (experiments); n = 2 (papers)), but impaired glucose control in glucose tolerance tests (SMD = -1.30 [-1.96, -0.63]; n = 6 (experiments); n = 3 (papers)). The impaired glycemic control following perfluoro-octane sulphonic acid (PFOS) exposure and enhanced mass loss following dichlorodiphenyltrichloroethane (DDT) exposure have not been replicated. Animal studies thus suggest some chemical groups, especially PCB and PFOS, could impair glucose control management during calorie restriction, similar to conclusions from limited existing clinical studies. We discuss the research that is urgently required to inform weight management services that are now the mainstay prevention initiative for T2DM.
Collapse
Affiliation(s)
- K A Bennett
- Division of Health Sciences, School of Applied Sciences, Kydd Building, Abertay University, Dundee, DD1 1HG.
| | | | - A L Savage
- Division of Health Sciences, School of Applied Sciences, Kydd Building, Abertay University, Dundee, DD1 1HG
| |
Collapse
|
3
|
Sun P, Guo X, Ding E, Li C, Ren H, Xu Y, Qian J, Deng F, Shi W, Dong H, Lin EZ, Guo P, Fang J, Zhang Q, Zhao W, Tong S, Lu X, Pollitt KJG, Shi X, Tang S. Association between Personal Abiotic Airborne Exposures and Body Composition Changes among Healthy Adults (60-69 Years Old): A Combined Exposome-Wide and Lipidome Mediation Approach from the China BAPE Study. ENVIRONMENTAL HEALTH PERSPECTIVES 2024; 132:77005. [PMID: 39028628 PMCID: PMC11259245 DOI: 10.1289/ehp13865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 05/25/2024] [Accepted: 06/24/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUND Evidence suggested that abiotic airborne exposures may be associated with changes in body composition. However, more evidence is needed to identify key pollutants linked to adverse health effects and their underlying biomolecular mechanisms, particularly in sensitive older adults. OBJECTIVES Our research aimed to systematically assess the relationship between abiotic airborne exposures and changes in body composition among healthy older adults, as well as the potential mediating mechanisms through the serum lipidome. METHODS From September 2018 to January 2019, we conducted a monthly survey among 76 healthy adults (60-69 years old) in the China Biomarkers of Air Pollutant Exposure (BAPE) study, measuring their personal exposures to 632 abiotic airborne pollutions using MicroPEM and the Fresh Air wristband, 18 body composition indicators from the InBody 770 device, and lipidomics from venous blood samples. We used an exposome-wide association study (ExWAS) and deletion/substitution/addition (DSA) model to unravel complex associations between exposure to contaminant mixtures and body composition, a Bayesian kernel machine regression (BKMR) model to assess the overall effect of key exposures on body composition, and mediation analysis to identify lipid intermediators. RESULTS The ExWAS and DSA model identified that 2,4,5-T methyl ester (2,4,5-TME), 9,10-Anthracenedione (ATQ), 4b,8-dimethyl-2-isopropylphenanthrene, and 4b,5,6,7,8,8a,9,10-octahydro-(DMIP) were associated with increased body fat mass (BFM), fat mass indicators (FMI), percent body fat (PBF), and visceral fat area (VFA) in healthy older adults [Bonferroni-Hochberg false discovery rate ( FD R BH ) < 0.05 ]. The BKMR model demonstrated a positive correlation between contaminants (anthracene, ATQ, copaene, di-epi-α -cedrene, and DMIP) with VFA. Mediation analysis revealed that phosphatidylcholine [PC, PC(16:1e/18:1), PC(16:2e/18:0)] and sphingolipid [SM, SM(d18:2/24:1)] mediated a significant portion, ranging from 12.27% to 26.03% (p-value < 0.05 ), of the observed increase in VFA. DISCUSSION Based on the evidence from multiple model results, ATQ and DMIP were statistically significantly associated with the increased VFA levels of healthy older adults, potentially regulated through lipid intermediators. These findings may have important implications for identifying potentially harmful environmental chemicals and developing targeted strategies for the control and prevention of chronic diseases in the future, particularly as the global population is rapidly aging. https://doi.org/10.1289/EHP13865.
Collapse
Affiliation(s)
- Peijie Sun
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Xiaojie Guo
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| | - Enmin Ding
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
- School of Population Medicine and Public Health, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chenfeng Li
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
- Department of Occupational Health and Environment Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Huimin Ren
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Yibo Xu
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
- Department of Epidemiology, School of Public Health, China Medical University, Shenyang, China
| | - Jiankun Qian
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Fuchang Deng
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Wanying Shi
- Department of Epidemiology and Health Statistics, School of Public Health, Capital Medical University, and Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, China
| | - Haoran Dong
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Elizabeth Z. Lin
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, Connecticut, USA
| | - Pengfei Guo
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, Connecticut, USA
| | - Jianlong Fang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Qian Zhang
- Chinese Center for Disease Control and Prevention, National Institute for Nutrition and Health, Beijing, China
| | - Wenhua Zhao
- Chinese Center for Disease Control and Prevention, National Institute for Nutrition and Health, Beijing, China
| | - Shilu Tong
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
- School of Public Health and Social Work, Queensland University of Technology, Brisbane, Australia
| | - Xiaobo Lu
- Department of Toxicology, School of Public Health, China Medical University, Shenyang, China
| | - Krystal J. Godri Pollitt
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, Connecticut, USA
| | - Xiaoming Shi
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Song Tang
- China CDC Key Laboratory of Environment and Population Health, National Institute of Environmental Health, Chinese Center for Disease Control and Prevention, Beijing, China
- Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
4
|
Heianza Y, Xue Q, Rood J, Clish CB, Bray GA, Sacks FM, Qi L. Changes in bile acid subtypes and improvements in lipid metabolism and atherosclerotic cardiovascular disease risk: the Preventing Overweight Using Novel Dietary Strategies (POUNDS Lost) trial. Am J Clin Nutr 2024; 119:1293-1300. [PMID: 38428740 PMCID: PMC11130658 DOI: 10.1016/j.ajcnut.2024.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 01/26/2024] [Accepted: 02/26/2024] [Indexed: 03/03/2024] Open
Abstract
BACKGROUND Distinct circulating bile acid (BA) subtypes may play roles in regulating lipid homeostasis and atherosclerosis. OBJECTIVES We investigated whether changes in circulating BA subtypes induced by weight-loss dietary interventions were associated with improved lipid profiles and atherosclerotic cardiovascular disease (ASCVD) risk estimates. METHODS This study included adults with overweight or obesity (n = 536) who participated in a randomized weight-loss dietary intervention trial. Circulating primary and secondary unconjugated BAs and their taurine-/glycine-conjugates were measured at baseline and 6 mo after the weight-loss diet intervention. The ASCVD risk estimates were calculated using the validated equations. RESULTS At baseline, higher concentrations of specific BA subtypes were related to higher concentrations of atherogenic very low-density lipoprotein lipid subtypes and ASCVD risk estimates. Weight-loss diet-induced decreases in primary BAs were related to larger reductions in triglycerides and total cholesterol [every 1 standard deviation (SD) decrease of glycocholate, glycochenodeoxycholate, or taurochenodeoxycholate was related to β (standard error) -3.3 (1.3), -3.4 (1.3), or -3.8 (1.3) mg/dL, respectively; PFDR < 0.05 for all]. Greater decreases in specific secondary BA subtypes were also associated with improved lipid metabolism at 6 mo; there was β -4.0 (1.1) mg/dL per 1-SD decrease of glycoursodeoxycholate (PFDR =0.003) for changes in low-density lipoprotein cholesterol. We found significant interactions (P-interaction < 0.05) between dietary fat intake and changes in BA subtypes on changes in ASCVD risk estimates; decreases in primary and secondary BAs (such as conjugated cholate or deoxycholate) were significantly associated with improved ASCVD risk after consuming a high-fat diet, but not after consuming a low-fat diet. CONCLUSIONS Decreases in distinct BA subtypes were associated with improved lipid profiles and ASCVD risk estimates, highlighting the importance of changes in circulating BA subtypes as significant factors linked to improved lipid metabolism and ASCVD risk estimates in response to weight-loss dietary interventions. Habitual dietary fat intake may modify the associations of changes in BAs with ASCVD risk. This trial was registered at clinicaltrials.gov as NCT00072995.
Collapse
Affiliation(s)
- Yoriko Heianza
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States.
| | - Qiaochu Xue
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States
| | - Jennifer Rood
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States
| | - Clary B Clish
- Metabolomics Platform, Broad Institute of MIT and Harvard, Cambridge, MA, United States
| | - George A Bray
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, United States
| | - Frank M Sacks
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA, United States; Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, United States.
| |
Collapse
|
5
|
Padua E, Caprio M, Feraco A, Camajani E, Gorini S, Armani A, Ruscello B, Bellia A, Strollo R, Lombardo M. The Impact of Diet and Physical Activity on Fat-to-Lean Mass Ratio. Nutrients 2023; 16:19. [PMID: 38201847 PMCID: PMC10780510 DOI: 10.3390/nu16010019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 12/16/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
In this retrospective study, we evaluated the efficacy of a personalised low-calorie Mediterranean Diet (MD) in promoting fat mass (FM) reduction while preserving fat-free mass (FFM). This study involved 100 Caucasian adults aged 18-65 years who followed a tailored low-calorie MD for two months. The total energy expenditure was assessed using a multi-sensor armband. The change in body composition (BC) was evaluated using the Δ% FM-to-FFM ratio, calculated as the difference in the FM to FFM ratio before and after the diet, divided by the ratio before the diet, and multiplied by 100. A negative value indicates a greater decrease in FM than FFM, while a positive value suggests a greater increase in FM than FFM. This study demonstrated a significant FM reduction, with an average decrease of 5% (p < 0.001). However, the relationship between caloric reduction and the Δ% FM-to-FFM ratio showed a weak negative correlation (r = -0.03, p > 0.05). This suggests that the calorie deficit had a minimal direct impact on the BC changes. Subjects over the age of 30 showed an increase in muscle mass, while younger subjects showed no significant changes. Moreover, a direct correlation was observed between the changes in MET (Metabolic Equivalent of Task) values and the Δ% FM-to-FFM ratio, indicating that improved average physical activity intensity positively influences BC. In the female subgroup, high protein intake, exercise intensity, and the duration of physical activity were positively correlated with an improvement in the Δ% FM-to-FFM ratio. However, for individuals with BMI 20-25 kg/m2, high fibre intake was surprisingly negatively correlated with the Δ% FM-to-FFM ratio. This study underscores the intricate interplay between calorie restriction, physical activity intensity, and BC changes. It also suggests that individual factors, including age, gender, and BMI, may influence the response to a low-calorie MD. However, further prospective studies with larger sample sizes are necessary to confirm and expand upon these findings.
Collapse
Affiliation(s)
- Elvira Padua
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Open University, Via di Val Cannuta, 247, 00166 Rome, Italy; (E.P.); (M.C.); (A.F.); (E.C.); (S.G.); (A.A.); (B.R.); (R.S.)
| | - Massimiliano Caprio
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Open University, Via di Val Cannuta, 247, 00166 Rome, Italy; (E.P.); (M.C.); (A.F.); (E.C.); (S.G.); (A.A.); (B.R.); (R.S.)
- Laboratory of Cardiovascular Endocrinology, San Raffaele Research Institute, IRCCS San Raffaele Roma, Via di Val Cannuta, 247, 00166 Rome, Italy
| | - Alessandra Feraco
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Open University, Via di Val Cannuta, 247, 00166 Rome, Italy; (E.P.); (M.C.); (A.F.); (E.C.); (S.G.); (A.A.); (B.R.); (R.S.)
- Laboratory of Cardiovascular Endocrinology, San Raffaele Research Institute, IRCCS San Raffaele Roma, Via di Val Cannuta, 247, 00166 Rome, Italy
| | - Elisabetta Camajani
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Open University, Via di Val Cannuta, 247, 00166 Rome, Italy; (E.P.); (M.C.); (A.F.); (E.C.); (S.G.); (A.A.); (B.R.); (R.S.)
- Laboratory of Cardiovascular Endocrinology, San Raffaele Research Institute, IRCCS San Raffaele Roma, Via di Val Cannuta, 247, 00166 Rome, Italy
| | - Stefania Gorini
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Open University, Via di Val Cannuta, 247, 00166 Rome, Italy; (E.P.); (M.C.); (A.F.); (E.C.); (S.G.); (A.A.); (B.R.); (R.S.)
- Laboratory of Cardiovascular Endocrinology, San Raffaele Research Institute, IRCCS San Raffaele Roma, Via di Val Cannuta, 247, 00166 Rome, Italy
| | - Andrea Armani
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Open University, Via di Val Cannuta, 247, 00166 Rome, Italy; (E.P.); (M.C.); (A.F.); (E.C.); (S.G.); (A.A.); (B.R.); (R.S.)
- Laboratory of Cardiovascular Endocrinology, San Raffaele Research Institute, IRCCS San Raffaele Roma, Via di Val Cannuta, 247, 00166 Rome, Italy
| | - Bruno Ruscello
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Open University, Via di Val Cannuta, 247, 00166 Rome, Italy; (E.P.); (M.C.); (A.F.); (E.C.); (S.G.); (A.A.); (B.R.); (R.S.)
| | - Alfonso Bellia
- Department of Systems Medicine, University of Rome “Tor Vergata”, Via Montpellier 1, 00133 Rome, Italy;
| | - Rocky Strollo
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Open University, Via di Val Cannuta, 247, 00166 Rome, Italy; (E.P.); (M.C.); (A.F.); (E.C.); (S.G.); (A.A.); (B.R.); (R.S.)
| | - Mauro Lombardo
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Open University, Via di Val Cannuta, 247, 00166 Rome, Italy; (E.P.); (M.C.); (A.F.); (E.C.); (S.G.); (A.A.); (B.R.); (R.S.)
| |
Collapse
|
6
|
Li X, Shao X, Kou M, Wang X, Ma H, Grundberg E, Bazzano LA, Smith SR, Bray GA, Sacks FM, Qi L. DNA Methylation at ABCG1 and Long-term Changes in Adiposity and Fat Distribution in Response to Dietary Interventions: The POUNDS Lost Trial. Diabetes Care 2023; 46:2201-2207. [PMID: 37770056 PMCID: PMC10698224 DOI: 10.2337/dc23-0748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 09/07/2023] [Indexed: 10/03/2023]
Abstract
OBJECTIVE To examine whether participants with different levels of diabetes-related DNA methylation at ABCG1 might respond differently to dietary weight loss interventions with long-term changes in adiposity and body fat distribution. RESEARCH DESIGN AND METHODS The current study included overweight/obese participants from the POUNDS Lost trial. Blood levels of regional DNA methylation at ABCG1 were profiled by high-resolution methylC-capture sequencing at baseline among 673 participants, of whom 598 were followed up at 6 months and 543 at 2 years. Two-year changes in adiposity and computed tomography-measured body fat distribution were calculated. RESULTS Regional DNA methylation at ABCG1 showed significantly different associations with long-term changes in body weight and waist circumference at 6 months and 2 years in dietary interventions varying in protein intake (interaction P < 0.05 for all). Among participants assigned to an average-protein (15%) diet, lower baseline regional DNA methylation at ABCG1 was associated with greater reductions in body weight and waist circumference at 6 months and 2 years, whereas opposite associations were found among those assigned to a high-protein (25%) diet. Similar interaction patterns were also observed for body fat distribution, including visceral adipose tissue, subcutaneous adipose tissue, deep subcutaneous adipose tissue, and total adipose tissue at 6 months and 2 years (interaction P < 0.05 for all). CONCLUSIONS Baseline DNA methylation at ABCG1 interacted with dietary protein intake on long-term decreases in adiposity and body fat distribution. Participants with lower methylation at ABCG1 benefitted more in long-term reductions in body weight, waist circumference, and body fat distribution when consuming an average-protein diet.
Collapse
Affiliation(s)
- Xiang Li
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA
| | - Xiaojian Shao
- Digital Technologies Research Centre, National Research Council Canada, Ottawa, Ontario, Canada
| | - Minghao Kou
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA
| | - Xuan Wang
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA
| | - Hao Ma
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA
| | - Elin Grundberg
- Department of Pediatrics, Genomic Medicine Center, Children’s Mercy Kansas City, Kansas City, MO
| | - Lydia A. Bazzano
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA
| | | | - George A. Bray
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA
| | - Frank M. Sacks
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
7
|
Qi L, Heianza Y, Li X, Sacks FM, Bray GA. Toward Precision Weight-Loss Dietary Interventions: Findings from the POUNDS Lost Trial. Nutrients 2023; 15:3665. [PMID: 37630855 PMCID: PMC10458797 DOI: 10.3390/nu15163665] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/13/2023] [Accepted: 08/18/2023] [Indexed: 08/27/2023] Open
Abstract
The POUNDS Lost trial is a 2-year clinical trial testing the effects of dietary interventions on weight loss. This study included 811 adults with overweight or obesity who were randomized to one of four diets that contained either 15% or 25% protein and 20% or 40% fat in a 2 × 2 factorial design. By 2 years, participants on average lost from 2.9 to 3.6 kg in body weight in the four intervention arms, while no significant difference was observed across the intervention arms. In POUNDS Lost, we performed a series of ancillary studies to detect intrinsic factors particular to genomic, epigenomic, and metabolomic markers that may modulate changes in weight and other cardiometabolic traits in response to the weight-loss dietary interventions. Genomic variants identified from genome-wide association studies (GWASs) on obesity, type 2 diabetes, glucose and lipid metabolisms, gut microbiome, and dietary intakes have been found to interact with dietary macronutrients (fat, protein, and carbohydrates) in relation to weight loss and changes of body composition and cardiometabolic traits. In addition, we recently investigated epigenomic modifications, particularly blood DNA methylation and circulating microRNAs (miRNAs). We reported DNA methylation levels at NFATC2IP, CPT1A, TXNIP, and LINC00319 were related to weight loss or changes of glucose, lipids, and blood pressure; we also reported thrifty miRNA expression as a significant epigenomic marker related to changes in insulin sensitivity and adiposity. Our studies have also highlighted the importance of temporal changes in novel metabolomic signatures for gut microbiota, bile acids, and amino acids as predictors for achievement of successful weight loss outcomes. Moreover, our studies indicate that biochemical, behavioral, and psychosocial factors such as physical activity, sleep disturbance, and appetite may also modulate metabolic changes during dietary interventions. This review summarized our major findings in the POUNDS Lost trial, which provided preliminary evidence supporting the development of precision diet interventions for obesity management.
Collapse
Affiliation(s)
- Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70118, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Yoriko Heianza
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70118, USA
| | - Xiang Li
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70118, USA
| | - Frank M. Sacks
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - George A. Bray
- Department of Clinical Obesity, Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, USA
| |
Collapse
|
8
|
Dent R, Harris N, van Walraven C. Validity of two weight prediction models for community-living patients participating in a weight loss program. Sci Rep 2023; 13:11629. [PMID: 37468655 DOI: 10.1038/s41598-023-38683-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 07/12/2023] [Indexed: 07/21/2023] Open
Abstract
Models predicting individual body weights over time clarify patient expectations in weight loss programs. The accuracy of two commonly used weight prediction models in community living people is unclear. All eligible people entering a weight management program between 1992 and 2015 were included. Patients' diet was 1200 kcal/day for week 0 followed by 900 kcal/day for weeks 1-7 and were excluded from the analysis if they were nonadherent. We generated expected weights using the National Institutes of Health Body Weight Planner (NIH-BWP) and the Pennington Biomedical Research Center Weight Loss Predictor (PBRC-WLP). 3703 adherent people were included (mean age 46 years, 72.6% women, mean [SD] weight 262.3 pounds [54.2], mean [SD] BMI 42.4 [7.6]). Mean (SD) relative body weight differences (100*[observed-expected]/expected) for NIH-BWP and PBRC-WLP models was - 1.5% (3.8) and - 2.9% (3.2), respectively. At week 7, mean squared error with NIH-BWP (98.8, 83%CI 89.7-108.8) was significantly lower than that with PBRC-WLP (117.7, 83%CI 112.4-123.4). Notable variation in relative weight difference were seen (for NIH-BWP, 5th-95th percentile was - 6.2%, + 3.7%; Δ 9.9%). During the first 7 weeks of a weight loss program, both weight prediction models returned expected weights that were very close to observed values with the NIH-BWP being more accurate. However, notable variability between expected and observed weights in individual patients were seen. Clinicians can monitor patients in weight loss programs by comparing their progress with these data.
Collapse
Affiliation(s)
- Robert Dent
- Department of Medicine, The Ottawa Hospital, Ottawa, Canada
| | - Neil Harris
- Weight Management Clinic, The Ottawa Hospital, Ottawa, Canada
| | - Carl van Walraven
- Ottawa Hospital Research Institute, Institute for Clinical Evaluative Sciences, University of Ottawa, ASB1-003 1053, Carling Ave, Ottawa, ON, K1Y 4E9, Canada.
| |
Collapse
|
9
|
Cifuentes L, Ghusn W, Feris F, Campos A, Sacoto D, De la Rosa A, McRae A, Rieck T, Mansfield S, Ewoldt J, Friend J, Grothe K, Lennon RJ, Hurtado MD, Clark MM, Camilleri M, Hensrud DD, Acosta A. Phenotype tailored lifestyle intervention on weight loss and cardiometabolic risk factors in adults with obesity: a single-centre, non-randomised, proof-of-concept study. EClinicalMedicine 2023; 58:101923. [PMID: 37007741 PMCID: PMC10050763 DOI: 10.1016/j.eclinm.2023.101923] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 04/04/2023] Open
Abstract
Background Lifestyle interventions for weight loss are currently not individualised to underlying pathophysiology and behavioral traits in obesity. We aim to compare the outcome of a standard lifestyle intervention (SLI) to phenotype-tailored lifestyle interventions (PLI) on weight loss, cardiometabolic risk factors and physiologic variables contributing to obesity. Methods This 12-week, single-centre non-randomised proof-of-concept clinical trial including men and women aged 18-65 years with a body mass index (BMI) greater than 30 without history of any bariatric procedure, and current use of any medication known to affect weight. Participants lived anywhere in the United States, and underwent in-person testing in Rochester, MN at a teaching hospital. All participants completed in-person phenotype testing at baseline and after 12 weeks. Participants were assigned to their intervention based on their period of enrollment. In the first phase, participants were assigned to SLI with a low-calorie diet (LCD), moderate physical activity, and weekly behavioral therapy sessions. In the second phase, other participants were assigned to PLI according to phenotype: abnormal satiation (time-restricted volumetric LCD); abnormal postprandial satiety (LCD with pre-meal protein supplementation); emotional eating (LCD with intensive behavioral therapy); and abnormal resting energy expenditure (LCD with post-workout protein supplementation and high-intensity interval training). The primary outcome was total body weight loss in kg at 12 weeks using multiple imputation for missing data. Linear models estimated the association of study group allocation and study endpoints adjusting for age, sex, and baseline weight. This study was registered with ClinicalTrials.gov, NCT04073394. Findings Between July 2020 and August 2021, 211 participants were screened, and 165 were assigned to one of the two treatments in the two phases: 81 SLI (mean [SD] age 42.9 [12] years; 79% women; BMI 38.0 [6.0]) and 84 PLI (age 44.8 [12.2] years; 83% women; BMI 38.7 [6.9]); 146 completed the 12-week programs. The weight loss was -7.4 kg (95%CI, -8.8, -6.0) with PLI vs. -4.3 kg (95%CI, -5.8, -2.7) with SLI (difference, -3.1 kg [95%CI, -5.1 to -1.1]; P = 0.004). No adverse events were reported in any group. Interpretation Phenotype-tailored lifestyle interventions may result in significant weight loss, but a randomised controlled trial is required to confirm causality. Funding Mayo Clinic; NIH (K23-DK114460).
Collapse
Affiliation(s)
- Lizeth Cifuentes
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Wissam Ghusn
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Fauzi Feris
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Alejandro Campos
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Daniel Sacoto
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Alan De la Rosa
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Alison McRae
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Thom Rieck
- Dan Abraham Healthy Living Center, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Sara Mansfield
- Dan Abraham Healthy Living Center, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Jason Ewoldt
- Dan Abraham Healthy Living Center, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Jamie Friend
- Dan Abraham Healthy Living Center, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Karen Grothe
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition; Department of Medicine, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
- Department of Psychiatry & Psychology, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Ryan J. Lennon
- Department of Quantitative Health Sciences, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Maria D. Hurtado
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition; Department of Medicine, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Matthew M. Clark
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
- Division of Endocrinology, Diabetes, Metabolism, and Nutrition; Department of Medicine, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
- Division of General Internal Medicine, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Michael Camilleri
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Donald D. Hensrud
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
- Dan Abraham Healthy Living Center, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
- Division of General Internal Medicine, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| | - Andres Acosta
- Precision Medicine for Obesity Program, Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic and Mayo Foundation, Rochester, MN, USA
| |
Collapse
|
10
|
Igudesman D, Crandell J, Corbin KD, Zaharieva DP, Addala A, Thomas JM, Casu A, Kirkman MS, Pokaprakarn T, Riddell MC, Burger K, Pratley RE, Kosorok MR, Maahs DM, Mayer-Davis EJ. Weight management in young adults with type 1 diabetes: The advancing care for type 1 diabetes and obesity network sequential multiple assignment randomized trial pilot results. Diabetes Obes Metab 2023; 25:688-699. [PMID: 36314293 PMCID: PMC9898100 DOI: 10.1111/dom.14911] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 10/22/2022] [Accepted: 10/28/2022] [Indexed: 02/06/2023]
Abstract
AIMS Co-management of weight and glycaemia is critical yet challenging in type 1 diabetes (T1D). We evaluated the effect of a hypocaloric low carbohydrate, hypocaloric moderate low fat, and Mediterranean diet without calorie restriction on weight and glycaemia in young adults with T1D and overweight or obesity. MATERIALS AND METHODS We implemented a 9-month Sequential, Multiple Assignment, Randomized Trial pilot among adults aged 19-30 years with T1D for ≥1 year and body mass index 27-39.9 kg/m2 . Re-randomization occurred at 3 and 6 months if the assigned diet was not acceptable or not effective. We report results from the initial 3-month diet period and re-randomization statistics before shutdowns due to COVID-19 for primary [weight, haemoglobin A1c (HbA1c), percentage of time below range <70 mg/dl] and secondary outcomes [body fat percentage, percentage of time in range (70-180 mg/dl), and percentage of time below range <54 mg/dl]. Models adjusted for design, demographic and clinical covariates tested changes in outcomes and diet differences. RESULTS Adjusted weight and HbA1c (n = 38) changed by -2.7 kg (95% CI -3.8, -1.5, P < .0001) and -0.91 percentage points (95% CI -1.5, -0.30, P = .005), respectively, while adjusted body fat percentage remained stable, on average (P = .21). Hypoglycaemia indices remained unchanged following adjustment (n = 28, P > .05). Variability in all outcomes, including weight change, was considerable (57.9% were re-randomized primarily due to loss of <2% body weight). No outcomes varied by diet. CONCLUSIONS Three months of a diet, irrespective of macronutrient distribution or caloric restriction, resulted in weight loss while improving or maintaining HbA1c levels without increasing hypoglycaemia in adults with T1D.
Collapse
Affiliation(s)
- Daria Igudesman
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- AdventHealth Translational Research Institute, Orlando, FL 32804
| | - Jamie Crandell
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Karen D. Corbin
- AdventHealth Translational Research Institute, Orlando, FL 32804
| | - Dessi P. Zaharieva
- Department of Pediatrics, Division of Endocrinology, Stanford University, Stanford, CA 94304
| | - Ananta Addala
- Department of Pediatrics, Division of Endocrinology, Stanford University, Stanford, CA 94304
| | - Joan M. Thomas
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Anna Casu
- AdventHealth Translational Research Institute, Orlando, FL 32804
| | - M. Sue Kirkman
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Teeranan Pokaprakarn
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - Michael C. Riddell
- School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - Kyle Burger
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | | | - Michael R. Kosorok
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | - David M. Maahs
- Department of Pediatrics, Division of Endocrinology, Stanford University, Stanford, CA 94304
| | - Elizabeth J. Mayer-Davis
- Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
- Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599
| | | |
Collapse
|
11
|
Qi L. Nutrition for precision health: The time is now. Obesity (Silver Spring) 2022; 30:1335-1344. [PMID: 35785484 DOI: 10.1002/oby.23448] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/13/2022] [Accepted: 03/21/2022] [Indexed: 11/11/2022]
Abstract
Precision nutrition has emerged as a boiling area of nutrition research, with a particular focus on revealing the individual variability in response to diets that is determined mainly by the complex interactions of dietary factors with the multi-tiered "omics" makeups. Reproducible findings from the observational studies and diet intervention trials have lent preliminary but consistent evidence to support the fundamental role of gene-diet interactions in determining the individual variability in health outcomes including obesity and weight loss. Recent investigations suggest that the abundance and diversity of the gut microbiome may also modify the dietary effects; however, considerable instability in the results from the microbiome research has been noted. In addition, growing studies suggest that a complicated multiomics algorithm would be developed by incorporating the genome, epigenome, metabolome, proteome, and microbiome in predicting the individual variability in response to diets. Moreover, precision nutrition would also scrutinize the role of biological (circadian) rhythm in determining the individual variability of dietary effects. The evidence gathered from precision nutrition research will be the basis for constructing precision health dietary recommendations, which hold great promise to help individuals and their health care providers create precise and effective diet plans for precision health in the future.
Collapse
Affiliation(s)
- Lu Qi
- Department of Epidemiology, School of Public Health and Tropical Medicine, Tulane University, New Orleans, Louisiana, USA
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Gkouskou KK, Grammatikopoulou MG, Lazou E, Sanoudou D, Goulis DG, Eliopoulos AG. Genetically-Guided Medical Nutrition Therapy in Type 2 Diabetes Mellitus and Pre-diabetes: A Series of n-of-1 Superiority Trials. Front Nutr 2022; 9:772243. [PMID: 35265654 PMCID: PMC8899711 DOI: 10.3389/fnut.2022.772243] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Accepted: 01/12/2022] [Indexed: 12/12/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is a heterogeneous metabolic disorder of multifactorial etiology that includes genetic and dietary influences. By addressing the latter, medical nutrition therapy (MNT) contributes to the management of T2DM or pre-diabetes toward achieving glycaemic control and improved insulin sensitivity. However, the clinical outcomes of MNT vary and may further benefit from personalized nutritional plans that take into consideration genetic variations associated with individual responses to macronutrients. The aim of the present series of n-of-1 trials was to assess the effects of genetically-guided vs. conventional MNT on patients with pre-diabetes or T2DM. A quasi-experimental, cross-over design was adopted in three Caucasian adult men with either diagnosis. Complete diet, bioclinical and anthropometric assessment was performed and a conventional MNT, based on the clinical practice guidelines was applied for 8 weeks. After a week of “wash-out,” a precision MNT was prescribed for an additional 8-week period, based on the genetic characteristics of each patient. Outcomes of interest included changes in body weight (BW), fasting plasma glucose (FPG), and blood pressure (BP). Collectively, the trials indicated improvements in BW, FPG, BP, and glycosylated hemoglobin (HbA1c) following the genetically-guided precision MNT intervention. Moreover, both patients with pre-diabetes experienced remission of the condition. We conclude that improved BW loss and glycemic control can be achieved in patients with pre-diabetes/T2DM, by coupling MNT to their genetic makeup, guiding optimal diet, macronutrient composition, exercise and oral nutrient supplementation in a personalized manner.
Collapse
Affiliation(s)
- Kalliopi K Gkouskou
- Department of Biology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Embiodiagnostics Biology Research Company, Heraklion, Greece
| | - Maria G Grammatikopoulou
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, Faculty of Health Sciences, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece.,Department of Nutritional Sciences and Dietetics, Faculty of Health Sciences, International Hellenic University, Thessaloniki, Greece
| | - Evgenia Lazou
- Department of Biology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Despina Sanoudou
- Clinical Genomics and Pharmacogenomics Unit, Fourth Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Dimitrios G Goulis
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, Faculty of Health Sciences, Medical School, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Aristides G Eliopoulos
- Department of Biology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.,Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
13
|
CYP1A2 polymorphisms modify the association of habitual coffee consumption with appetite, macronutrient intake, and body mass index: results from an observational cohort and a cross-over randomized study. Int J Obes (Lond) 2021; 46:162-168. [PMID: 34564706 DOI: 10.1038/s41366-021-00972-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 09/07/2021] [Accepted: 09/15/2021] [Indexed: 01/14/2023]
Abstract
BACKGROUND/OBJECTIVES Evidence regarding the influence of coffee on appetite and weight control is equivocal and the influence of covariates, such as genetic variation in caffeine metabolism, remains unknown. Herein, we addressed the novel hypothesis that genetic variation in CYP1A2, a gene responsible for more than 95% of caffeine metabolism, differentially impacts the association of coffee consumption with appetite and BMI among individuals with different genetic predispositions to obesity. SUBJECTS/METHODS A cross-over randomized intervention study involving 18 volunteers assessed the effects of coffee consumption on dietary intake, appetite, and levels of the appetite-controlling hormones asprosin and leptin. Data on habitual coffee intake, BMI, and perceived appetite were obtained from an observational cohort of 284 volunteers using validated questionnaires. Participants were stratified according to a validated genetic risk score (GRS) for obesity and to the -163C > A (rs762551) polymorphism of CYP1A2 as rapid (AA), intermediate (AC), or slow (CC) caffeine metabolizers. RESULTS Coffee consumption led to lower energy and dietary fat intake and circulating asprosin levels (P for interaction of rs762551 genotype*coffee consumption=0.056, 0.039, and 0.043, respectively) as compared to slow/intermediate metabolizers. High coffee consumption was more prevalent in rapid compared to slow metabolizers (P = 0.008 after adjustment for age, sex, and BMI) and was associated with lower appetite perception and lower BMI only in rapid metabolizers (P for interaction of rs762551 genotype*coffee consumption = 0.002 and 0.048, respectively). This differential association of rs762551 genotype and coffee consumption with BMI was more evident in individuals at higher genetic risk of obesity (mean adjusted difference in BMI = -5.82 kg/m2 for rapid versus slow/intermediate metabolizers who consumed more than 14 cups of coffee per week). CONCLUSIONS CYP1A2 rs762551 polymorphism modifies the association of habitual coffee consumption with BMI, in part by influencing appetite, energy intake and circulating levels of the orexigenic hormone asprosin. This association is more evident in subjects with high genetic predisposition to obesity. ClinicalTrials.gov: registered Clinical Trial NCT04514588.
Collapse
|
14
|
Whytock KL, Corbin KD, Parsons SA, Pachori A, Bock CP, Jones KP, Smith JS, Yi F, Xie H, Petucci CJ, Gardell SJ, Smith SR. Metabolic adaptation characterizes short-term resistance to weight loss induced by a low-calorie diet in overweight/obese individuals. Am J Clin Nutr 2021; 114:267-280. [PMID: 33826697 DOI: 10.1093/ajcn/nqab027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 01/28/2021] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Low-calorie diet (LCD)-induced weight loss demonstrates response heterogeneity. Physiologically, a decrease in energy expenditure lower than what is predicted based on body composition (metabolic adaptation) and/or an impaired capacity to increase fat oxidation may hinder weight loss. Understanding the metabolic components that characterize weight loss success is important for optimizing weight loss strategies. OBJECTIVES We tested the hypothesis that overweight/obese individuals who had lower than expected weight loss in response to a 28-d LCD would be characterized by 1) impaired fat oxidation and 2) whole-body metabolic adaptation. We also characterized the molecular mechanisms associated with weight loss success/failure. METHODS This was a retrospective comparison of participants who met their predicted weight loss targets [overweight/obese diet sensitive (ODS), n = 23, females = 21, males = 2] and those that did not [overweight/obese diet resistant (ODR), n = 14, females = 12, males = 2] after a 28-d LCD (900-1000 kcal/d). We used whole-body (energy expenditure and fat oxidation) and tissue-specific measurements (metabolic proteins in skeletal muscle, gene expression in adipose tissue, and metabolites in serum) to detect metabolic properties and biomarkers associated with weight loss success. RESULTS The ODR group had greater mean ± SD metabolic adaptation (-175 ± 149 kcal/d; +119%) than the ODS group (-80 ± 108 kcal/d) after the LCD (P = 0.030). Mean ± SD fat oxidation increased similarly for both groups from baseline (0.0701 ± 0.0206 g/min) to day 28 (0.0869 ± 0.0269 g/min; P < 0.001). A principal component analysis factor comprised of serum 3-hydroxybutyric acid, citrate, leucine/isoleucine, acetyl-carnitine, and 3-hydroxylbutyrlcarnitine was associated with weight loss success at day 28 (std. β = 0.674, R2 = 0.479, P < 0.001). CONCLUSIONS Individuals who achieved predicted weight loss targets after a 28-d LCD were characterized by reduced metabolic adaptation. Accumulation of metabolites associated with acetyl-CoA excess and enhanced ketogenesis was identified in the ODS group.This trial was registered at clinicaltrials.gov as NCT01616082.
Collapse
Affiliation(s)
- Katie L Whytock
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Karen D Corbin
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | | | - Alok Pachori
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | | | - Karen P Jones
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Joshua S Smith
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Fanchao Yi
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Hui Xie
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Christopher J Petucci
- Translational Research Institute, AdventHealth, Orlando, FL, USA.,Cardiovascular Institute and Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | | | - Steven R Smith
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| |
Collapse
|
15
|
Gijbels A, Trouwborst I, Jardon KM, Hul GB, Siebelink E, Bowser SM, Yildiz D, Wanders L, Erdos B, Thijssen DHJ, Feskens EJM, Goossens GH, Afman LA, Blaak EE. The PERSonalized Glucose Optimization Through Nutritional Intervention (PERSON) Study: Rationale, Design and Preliminary Screening Results. Front Nutr 2021; 8:694568. [PMID: 34277687 PMCID: PMC8278004 DOI: 10.3389/fnut.2021.694568] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/03/2021] [Indexed: 12/13/2022] Open
Abstract
Background: It is well-established that the etiology of type 2 diabetes differs between individuals. Insulin resistance (IR) may develop in different tissues, but the severity of IR may differ in key metabolic organs such as the liver and skeletal muscle. Recent evidence suggests that these distinct tissue-specific IR phenotypes may also respond differentially to dietary macronutrient composition with respect to improvements in glucose metabolism. Objective: The main objective of the PERSON study is to investigate the effects of an optimal vs. suboptimal dietary macronutrient intervention according to tissue-specific IR phenotype on glucose metabolism and other health outcomes. Methods: In total, 240 overweight/obese (BMI 25 – 40 kg/m2) men and women (age 40 – 75 years) with either skeletal muscle insulin resistance (MIR) or liver insulin resistance (LIR) will participate in a two-center, randomized, double-blind, parallel, 12-week dietary intervention study. At screening, participants undergo a 7-point oral glucose tolerance test (OGTT) to determine the hepatic insulin resistance index (HIRI) and muscle insulin sensitivity index (MISI), classifying each participant as either “No MIR/LIR,” “MIR,” “LIR,” or “combined MIR/LIR.” Individuals with MIR or LIR are randomized to follow one of two isocaloric diets varying in macronutrient content and quality, that is hypothesized to be either an optimal or suboptimal diet, depending on their tissue-specific IR phenotype (MIR/LIR). Extensive measurements in a controlled laboratory setting as well as phenotyping in daily life are performed before and after the intervention. The primary study outcome is the difference in change in disposition index, which is the product of insulin sensitivity and first-phase insulin secretion, between participants who received their hypothesized optimal or suboptimal diet. Discussion: The PERSON study is one of the first randomized clinical trials in the field of precision nutrition to test effects of a more personalized dietary intervention based on IR phenotype. The results of the PERSON study will contribute knowledge on the effectiveness of targeted nutritional strategies to the emerging field of precision nutrition, and improve our understanding of the complex pathophysiology of whole body and tissue-specific IR. Clinical Trial Registration:https://clinicaltrials.gov/ct2/show/NCT03708419, clinicaltrials.gov as NCT03708419.
Collapse
Affiliation(s)
- Anouk Gijbels
- Division of Human Nutrition and Health, Wageningen University, Wageningen, Netherlands.,Top Institute Food and Nutrition, Wageningen, Netherlands
| | - Inez Trouwborst
- Top Institute Food and Nutrition, Wageningen, Netherlands.,Department of Human Biology, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Kelly M Jardon
- Top Institute Food and Nutrition, Wageningen, Netherlands.,Department of Human Biology, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Gabby B Hul
- Department of Human Biology, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Els Siebelink
- Division of Human Nutrition and Health, Wageningen University, Wageningen, Netherlands
| | - Suzanne M Bowser
- Department of Human Biology, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Dilemin Yildiz
- Department of Human Biology, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Lisa Wanders
- Top Institute Food and Nutrition, Wageningen, Netherlands.,Department of Physiology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, Netherlands
| | - Balázs Erdos
- Top Institute Food and Nutrition, Wageningen, Netherlands.,Maastricht Centre for Systems Biology, Maastricht University, Maastricht, Netherlands
| | - Dick H J Thijssen
- Department of Physiology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, Netherlands.,Research Institute for Sport and Exercise Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Edith J M Feskens
- Division of Human Nutrition and Health, Wageningen University, Wageningen, Netherlands
| | - Gijs H Goossens
- Department of Human Biology, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Lydia A Afman
- Division of Human Nutrition and Health, Wageningen University, Wageningen, Netherlands
| | - Ellen E Blaak
- Top Institute Food and Nutrition, Wageningen, Netherlands.,Department of Human Biology, Maastricht University Medical Center+, Maastricht, Netherlands
| |
Collapse
|
16
|
Bouchard C. Genetics of Obesity: What We Have Learned Over Decades of Research. Obesity (Silver Spring) 2021; 29:802-820. [PMID: 33899337 DOI: 10.1002/oby.23116] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/15/2020] [Accepted: 12/16/2020] [Indexed: 12/14/2022]
Abstract
There is a genetic component to human obesity that accounts for 40% to 50% of the variability in body weight status but that is lower among normal weight individuals (about 30%) and substantially higher in the subpopulation of individuals with obesity and severe obesity (about 60%-80%). The appreciation that heritability varies across classes of BMI represents an important advance. After controlling for BMI, ectopic fat and fat distribution traits are characterized by heritability levels ranging from 30% to 55%. Defects in at least 15 genes are the cause of monogenic obesity cases, resulting mostly from deficiencies in the leptin-melanocortin signaling pathway. Approximately two-thirds of the BMI heritability can be imputed to common DNA variants, whereas low-frequency and rare variants explain the remaining fraction. Diminishing allele effect size is observed as the number of obesity-associated variants expands, with most BMI-increasing or -decreasing alleles contributing only a few grams or less to body weight. Obesity-promoting alleles exert minimal effects in normal weight individuals but have larger effects in individuals with a proneness to obesity, suggesting a higher penetrance; however, it is not known whether these larger effect sizes precede obesity or are caused by an obese state. The obesity genetic risk is conditioned by thousands of DNA variants that make genetically based obesity prevention and treatment a major challenge.
Collapse
Affiliation(s)
- Claude Bouchard
- Pennington Biomedical Research Center, Baton Rouge, Louisiana, USA
| |
Collapse
|
17
|
Mehta M, Istfan NW, Apovian CM. Obesity: Overview of Weight Management. Endocr Pract 2021; 27:626-635. [PMID: 33901648 DOI: 10.1016/j.eprac.2021.04.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 04/02/2021] [Accepted: 04/05/2021] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Obesity is a chronic illness that requires a multifaceted personalized treatment approach. METHODS & FINDINGS Using current guidelines and recent studies in weight management, this article reviews the multiple components of weight management: lifestyle intervention (dietary intervention, physical activity, and behavioral interventions), pharmacotherapy, endoscopic procedures, and surgical procedures. This review briefly discusses specific diets and dietary strategies, compensatory mechanisms acting against weight loss, recent changes to Food and Drug Administration approved antiobesity medications, and technological advances in weight management delivery. CONCLUSION Current literature is lacking large studies on the safety and efficacy of combination therapies involving pharmacotherapy plus 1 or more procedures.
Collapse
Affiliation(s)
- Meetal Mehta
- Brigham and Women's Hospital, Center for Weight Management and Wellness, Section of Endocrinology, Diabetes and Hypertension, Harvard Medical School, Boston, Massachusetts.
| | - Nawfal W Istfan
- Brigham and Women's Hospital, Center for Weight Management and Wellness, Section of Endocrinology, Diabetes and Hypertension, Harvard Medical School, Boston, Massachusetts
| | - Caroline M Apovian
- Brigham and Women's Hospital, Center for Weight Management and Wellness, Section of Endocrinology, Diabetes and Hypertension, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
18
|
Loos RJF, Burant C, Schur EA. Strategies to Understand the Weight-Reduced State: Genetics and Brain Imaging. Obesity (Silver Spring) 2021; 29 Suppl 1:S39-S50. [PMID: 33759393 PMCID: PMC8500189 DOI: 10.1002/oby.23101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/03/2020] [Accepted: 12/03/2020] [Indexed: 11/09/2022]
Abstract
Most individuals with obesity or overweight have difficulty maintaining weight loss. The weight-reduced state induces changes in many physiological processes that appear to drive weight regain. Here, we review the use of cell biology, genetics, and imaging techniques that are being used to begin understanding why weight regain is the normal response to dieting. As with obesity itself, weight regain has both genetic and environmental drivers. Genetic drivers for "thinness" and "obesity" largely overlap, but there is evidence for specific genetic loci that are different for each of these weight states. There is only limited information regarding the genetics of weight regain. Currently, most genetic loci related to weight point to the central nervous system as the organ responsible for determining the weight set point. Neuroimaging tools have proved useful in studying the contribution of the central nervous system to the weight-reduced state in humans. Neuroimaging technologies fall into three broad categories: functional, connectivity, and structural neuroimaging. Connectivity and structural imaging techniques offer unique opportunities for testing mechanistic hypotheses about changes in brain function or tissue structure in the weight-reduced state.
Collapse
Affiliation(s)
- Ruth J. F. Loos
- The Charles Bronfman Institute for Personalized Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- The Mindich Child Health and Development Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Charles Burant
- Department of Internal Medicine, University of Washington, Seattle, Washington, USA
| | - Ellen A. Schur
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
19
|
Bray GA, Ryan DH. Evidence-based weight loss interventions: Individualized treatment options to maximize patient outcomes. Diabetes Obes Metab 2021; 23 Suppl 1:50-62. [PMID: 32969147 DOI: 10.1111/dom.14200] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/17/2020] [Accepted: 09/20/2020] [Indexed: 12/17/2022]
Abstract
Against the backdrop of obesity as a major public health problem, we examined three questions: How much weight loss is needed to benefit patients with obesity? How well do current therapies do in producing weight loss? What strategies can be used to improve patient outcomes using evidence-based studies. This paper reviews literature on the outcomes of lifestyle, diet, medications and surgical treatments for obesity using literature searches for obesity treatments. Current treatments, including lifestyle, diet and exercise, produce a weight loss of 5% to 7% on average. Despite continued attempts to identify superior dietary approaches, most careful comparisons find that low carbohydrate diets are not significantly better than low fat diets for weight loss. The four medications currently approved by the US Food and Drug Administration for long-term management of obesity are not as effective as surgery, adding about 5% on average to lifestyle approaches to weight loss. Two new medications that are under investigation, semaglutide and tirzepatide, significantly improve on this. For all treatments for weight loss, including lifestyle, medications and surgery, there is enormous variability in the amount of weight lost. Examination of this literature has yielded evidence supporting baseline and process predictors, but the effect sizes associated with these predictors are small and there are no prospective studies showing that a personalized approach based on genotype or phenotype will yield uniform success. Because obesity is a chronic disease it requires a 'continuous treatment model' across the lifespan.
Collapse
Affiliation(s)
- George A Bray
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana
| | - Donna H Ryan
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, Louisiana
| |
Collapse
|
20
|
Hwalla N, Jaafar Z. Dietary Management of Obesity: A Review of the Evidence. Diagnostics (Basel) 2020; 11:diagnostics11010024. [PMID: 33375554 PMCID: PMC7823549 DOI: 10.3390/diagnostics11010024] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 12/08/2020] [Accepted: 12/08/2020] [Indexed: 01/29/2023] Open
Abstract
Obesity is a multi-factorial disease and its prevention and management require knowledge of the complex interactions underlying it and adopting a whole system approach that addresses obesogenic environments within country specific contexts. The pathophysiology behind obesity involves a myriad of genetic, epigenetic, physiological, and macroenvironmental factors that drive food intake and appetite and increase the obesity risk for susceptible individuals. Metabolically, food intake and appetite are regulated via intricate processes and feedback systems between the brain, gastrointestinal system, adipose and endocrine tissues that aim to maintain body weight and energy homeostasis but are also responsive to environmental cues that may trigger overconsumption of food beyond homeostatic needs. Under restricted caloric intake conditions such as dieting, these processes elicit compensatory metabolic mechanisms that promote energy intake and weight regain, posing great challenges to diet adherence and weight loss attempts. To mitigate these responses and enhance diet adherence and weight loss, different dietary strategies have been suggested in the literature based on their differential effects on satiety and metabolism. In this review article, we offer an overview of the literature on obesity and its underlying pathological mechanisms, and we present an evidence based comparative analysis of the effects of different popular dietary strategies on weight loss, metabolic responses and diet adherence in obesity.
Collapse
|
21
|
Wagner MA, Wesmiller SW, Maydick M, Gawron LM, Peterson-Burch FM, Conley YP. Symptom Science: Omics and Response to Non-Pharmacological Interventions. Biol Res Nurs 2020; 23:394-401. [PMID: 33267608 DOI: 10.1177/1099800420975205] [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: 11/15/2022]
Abstract
Incorporating omics into non-pharmacological intervention research design could provide a better understanding of the variability in response to these interventions. It would also provide evidence for precision-based non-pharmacological interventions, including interventions focused on symptoms. The purpose of this manuscript was to present examples of studies that have used omics to examine response to non-pharmacological intervention. Using the interventions of exercise, diet (related to obesity), cognitive based therapy, and alternative mind-body practices (meditation, yoga, and tai chi), PubMed was searched to identify studies that incorporated genomic or other omic approaches as part of a non-pharmacological intervention. The review identified genes associated with the effectiveness of each of the interventions. Although there were no genes that were associated with all four interventions, there were nine genes that were the focus of more than one intervention (ACE, BDNF, COMT, CXCL8, IL6, SL6A4, TNF, GSTM1, PTGER3). All nine of these genes were either directly or indirectly biologically related to one another, suggesting that this cadre of genes could serve as an initiation point for investigations using omic approaches to better understand response to non-pharmacological interventions.
Collapse
Affiliation(s)
| | | | | | - Lisa M Gawron
- School of Nursing, 6614University of Pittsburgh, PA, USA
| | | | | |
Collapse
|
22
|
Dent R, McPherson R, Harper ME. Factors affecting weight loss variability in obesity. Metabolism 2020; 113:154388. [PMID: 33035570 DOI: 10.1016/j.metabol.2020.154388] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/19/2020] [Accepted: 09/23/2020] [Indexed: 12/25/2022]
Abstract
Current obesity treatment strategies include diet, exercise, bariatric surgery, and a limited but growing repertoire of medications. Individual weight loss in response to each of these strategies is highly variable. Here we review research into factors potentially contributing to inter-individual variability in response to treatments for obesity, with a focus on studies in humans. Well-recognized factors associated with weight loss capacity include diet adherence, physical activity, sex, age, and specific medications. However, following control for each of these, differences in weight loss appear to persist in response to behavioral, pharmacological and surgical interventions. Adaptation to energy deficit involves complex feedback mechanisms, and inter-individual differences likely to arise from a host of poorly defined genetic factors, as well as differential responses in neurohormonal mechanisms (including gastrointestinal peptides), metabolic efficiency and capacity of tissues, non-exercise activity thermogenesis, thermogenic response to food, and in gut microbiome. A better understanding of the factors involved in inter-individual variability in response to therapies will guide more personalized approaches to the treatment of obesity.
Collapse
Affiliation(s)
- Robert Dent
- Department of Medicine, Division of Endocrinology and The Ottawa Hospital, University of Ottawa, 210 Melrose Ave, Ottawa, ON K1Y 4K7, Canada
| | - Ruth McPherson
- Atherogenomics Laboratory, Division of Cardiology, University of Ottawa Heart Institute, 40 Ruskin St., Ottawa, ON K1Y 4W7, Canada
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, Ottawa Institute of Systems Biology, University of Ottawa, 451 Smyth Rd., Ottawa, ON K1H 8M5, Canada.
| |
Collapse
|
23
|
Magkos F, Hjorth MF, Astrup A. Diet and exercise in the prevention and treatment of type 2 diabetes mellitus. Nat Rev Endocrinol 2020; 16:545-555. [PMID: 32690918 DOI: 10.1038/s41574-020-0381-5] [Citation(s) in RCA: 213] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/12/2020] [Indexed: 02/08/2023]
Abstract
Evidence from observational studies and randomized trials suggests that prediabetes and type 2 diabetes mellitus (T2DM) can develop in genetically susceptible individuals in parallel with weight (that is, fat) gain. Accordingly, studies show that weight loss can produce remission of T2DM in a dose-dependent manner. A weight loss of ~15 kg, achieved by calorie restriction as part of an intensive management programme, can lead to remission of T2DM in ~80% of patients with obesity and T2DM. However, long-term weight loss maintenance is challenging. Obesity and T2DM are associated with diminished glucose uptake in the brain that impairs the satiating effect of dietary carbohydrate; therefore, carbohydrate restriction might help maintain weight loss and maximize metabolic benefits. Likewise, increases in physical activity and fitness are an important contributor to T2DM remission when combined with calorie restriction and weight loss. Preliminary studies suggest that a precision dietary management approach that uses pretreatment glycaemic status to stratify patients can help optimize dietary recommendations with respect to carbohydrate, fat and dietary fibre. This approach might lead to improved weight loss maintenance and glycaemic control. Future research should focus on better understanding the individual response to dietary treatment and translating these findings into clinical practice.
Collapse
Affiliation(s)
- Faidon Magkos
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg Campus, Copenhagen, Denmark
| | - Mads F Hjorth
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg Campus, Copenhagen, Denmark
| | - Arne Astrup
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Frederiksberg Campus, Copenhagen, Denmark.
| |
Collapse
|
24
|
Gkouskou K, Vlastos I, Karkalousos P, Chaniotis D, Sanoudou D, Eliopoulos AG. The "Virtual Digital Twins" Concept in Precision Nutrition. Adv Nutr 2020; 11:1405-1413. [PMID: 32770212 PMCID: PMC7666894 DOI: 10.1093/advances/nmaa089] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/15/2020] [Accepted: 07/06/2020] [Indexed: 12/13/2022] Open
Abstract
Nutritional and lifestyle changes remain at the core of healthy aging and disease prevention. Accumulating evidence underscores the impact of genetic, metabolic, and host gut microbial factors on individual responses to nutrients, paving the way for the stratification of nutritional guidelines. However, technological advances that incorporate biological, nutritional, lifestyle, and health data at an unprecedented scale and depth conceptualize a future where preventative dietary interventions will exceed stratification and will be highly individualized. We herein discuss how genetic information combined with longitudinal metabolomic, immune, behavioral, and gut microbial parameters, and bioclinical variables could define a digital replica of oneself, a "virtual digital twin," which could serve to guide nutrition in a personalized manner. Such a model may revolutionize the management of obesity and its comorbidities, and provide a pillar for healthy aging.
Collapse
Affiliation(s)
| | - Ioannis Vlastos
- Department of Biology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Petros Karkalousos
- Department of Biomedical Sciences, University of West Attica, Athens, Greece
| | - Dimitrios Chaniotis
- Department of Biomedical Sciences, University of West Attica, Athens, Greece
| | - Despina Sanoudou
- Clinical Genomics and Pharmacogenomics Unit, 4th Department of Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece,Center for New Biotechnologies and Precision Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece,Center of Basic Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | | |
Collapse
|
25
|
Robinson K, Rozga M, Braakhuis A, Ellis A, Monnard CR, Sinley R, Wanner A, Vargas AJ. Effect of Incorporating Genetic Testing Results into Nutrition Counseling and Care on Dietary Intake: An Evidence Analysis Center Systematic Review-Part I. J Acad Nutr Diet 2020; 121:553-581.e3. [PMID: 32624394 DOI: 10.1016/j.jand.2020.04.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Indexed: 12/15/2022]
Abstract
Consumer interest in personalized nutrition based on nutrigenetic testing is growing. Recently, multiple, randomized controlled trials have sought to understand whether incorporating genetic information into dietary counseling alters dietary outcomes. The objective of this systematic review was to examine how incorporating genetic information into nutrition counseling and care, compared to an alternative intervention or control group, impacts dietary outcomes. This is the first of a 2-part systematic review series. Part II reports anthropometric, biochemical, and disease-specific outcomes. Peer-reviewed randomized controlled trials were identified through a systematic literature search of multiple databases, screened for eligibility, and critically reviewed and synthesized. Conclusion statements were graded to determine quality of evidence for each dietary outcome reported. Reported outcomes include intake of total energy and macronutrients, micronutrients, foods, food groups, food components (added sugar, caffeine, and alcohol), and composite diet scores. Ten articles representing 8 unique randomized controlled trials met inclusion criteria. Of 15 conclusion statements (evidence grades: Weak to Moderate), 13 concluded there was no significant effect of incorporating genetic information into nutrition counseling/care on dietary outcomes. Limited data suggested that carriers of higher-risk gene variants were more likely than carriers of low-risk gene variants to significantly reduce intake of sodium and alcohol in response to nutrition counseling that incorporated genetic results. Included studies differed in quality, selected genetic variants, timing and intensity of intervention, sample size, dietary assessment tools, and population characteristics. Therefore, strong conclusions could not be drawn. Collaboration between the Academy of Nutrition and Dietetics and professional nutrigenetic societies would likely prove valuable in prioritizing which genetic variants and targeted nutrition messages have the most potential to alter dietary outcomes in a given patient subpopulation and, thus, should be the targets of future research.
Collapse
Affiliation(s)
- Katie Robinson
- Scientific and Medical Affairs, Abbott Nutrition, Columbus, OH
| | - Mary Rozga
- Academy of Nutrition and Dietetics, Evidence Analysis Center, Chicago, IL.
| | - Andrea Braakhuis
- Faculty of Medical and Health Science, Discipline of Nutrition, The University of Auckland, Grafton, Auckland, New Zealand
| | - Amy Ellis
- University of Alabama, Tuscaloosa, AL
| | | | | | | | - Ashley J Vargas
- National Institutes of Health, Office of Disease Prevention, Rockville, MD
| |
Collapse
|
26
|
Ellis A, Rozga M, Braakhuis A, Monnard CR, Robinson K, Sinley R, Wanner A, Vargas AJ. Effect of Incorporating Genetic Testing Results into Nutrition Counseling and Care on Health Outcomes: An Evidence Analysis Center Systematic Review-Part II. J Acad Nutr Diet 2020; 121:582-605.e17. [PMID: 32624396 DOI: 10.1016/j.jand.2020.02.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Indexed: 02/06/2023]
Abstract
In recent years, literature examining implementation of nutritional genomics into clinical practice has increased, including publication of several randomized controlled trials (RCTs). This systematic review addressed the following question: In children and adults, what is the effect of incorporating results of genetic testing into nutrition counseling and care compared with an alternative intervention or control group, on nutrition-related health outcomes? A literature search of MEDLINE, Embase, PsycINFO, CINAHL, and other databases was conducted for peer-reviewed RCTs published from January 2008 until December 2018. An international workgroup consisting of registered dietitian nutritionists, systematic review methodologists, and evidence analysts screened and reviewed articles, summarized data, conducted meta-analyses, and graded conclusion statements. The second in a two-part series, this article specifically summarizes evidence from RCTs that examined health outcomes (ie, quality of life, disease incidence and prevention of disease progression, or mortality), intermediate health outcomes (ie, anthropometric measures, body composition, or relevant laboratory measures routinely collected in practice), and adverse events as reported by study authors. Analysis of 11 articles from nine RCTs resulted in 16 graded conclusion statements. Among participants with nonalcoholic fatty liver disease, a diet tailored to genotype resulted in a greater reduction of percent body fat compared with a customary diet for nonalcoholic fatty liver disease. However, meta-analyses for the outcomes of total cholesterol, low-density lipoprotein cholesterol, body mass index, and weight yielded null results. Heterogeneity between studies and low certainty of evidence precluded development of strong conclusions about the incorporation of genetic information into nutrition practice. Although there are still relatively few well-designed RCTs to inform integration of genetic information into the Nutrition Care Process, the field of nutritional genomics is evolving rapidly, and gaps in the literature identified by this systematic review can inform future studies.
Collapse
|
27
|
Bray GA. In the Footsteps of Wilbur Olin Atwater: The Atwater Lecture for 2019. Adv Nutr 2020; 11:743-750. [PMID: 31925422 PMCID: PMC7231597 DOI: 10.1093/advances/nmz128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 10/31/2019] [Accepted: 11/08/2019] [Indexed: 11/12/2022] Open
Abstract
A central theme of Atwater's research was the development and application of methods to understand how human beings and animals adapt to the nutrients they ingest. The research described in this article also deals with adaptation to nutrition focusing on adaptation to overnutrition, adaptation to undernutrition, adaptation to dietary fat, adaptation to dietary protein, adaptation to micronutrients, and adaptation to sugar and high-fructose corn syrup (HFCS). Studies using overfeeding have shown several things. First, overfeeding did not change the thermic response to ingestion of food nor the coupling of oxidative phosphorylation in muscle to energy expended by muscles during work on a bicycle ergometer between 25 and 100 watts. Second, the response to overfeeding was significantly influenced by the quantity of protein in the diet. During carefully controlled studies of underfeeding of people with obesity, the macronutrient composition of the diet did not affect the magnitude of weight loss. However, baseline genetic and metabolic information could provide guidance for selecting among the lower or higher protein diets, and lower or higher fat diets. Adaptation to an increase in dietary fat from 35% to 50% is slow and variable in healthy sedentary men. Adaptation is more rapid and complete when these same men were physically active. This effect of muscular exercise was traced to changes in the metabolism of glucose in muscles where pathways inhibiting glucose metabolism were activated by exercise. Dietary patterns that increased the intake of calcium, magnesium, and potassium effectively lower blood pressure in individuals with high normal blood pressure. Finally, the intake of sugary beverages was related to the onset of the current epidemic of obesity.
Collapse
Affiliation(s)
- George A Bray
- Pennington Biomedical Research Center of Louisiana State University, and Children's Hospital of Oakland Research Institute (CHORI), Baton Rouge, LA and Oakland, CA, USA
| |
Collapse
|