1
|
Volek JS, Kackley ML, Buga A. Nutritional Considerations During Major Weight Loss Therapy: Focus on Optimal Protein and a Low-Carbohydrate Dietary Pattern. Curr Nutr Rep 2024; 13:422-443. [PMID: 38814519 PMCID: PMC11327213 DOI: 10.1007/s13668-024-00548-6] [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] [Accepted: 05/01/2024] [Indexed: 05/31/2024]
Abstract
PURPOSE OF REVIEW Considering the high prevalence of obesity and related metabolic impairments in the population, the unique role nutrition has in weight loss, reversing metabolic disorders, and maintaining health cannot be overstated. Normal weight and well-being are compatible with varying dietary patterns, but for the last half century there has been a strong emphasis on low-fat, low-saturated fat, high-carbohydrate based approaches. Whereas low-fat dietary patterns can be effective for a subset of individuals, we now have a population where the vast majority of adults have excess adiposity and some degree of metabolic impairment. We are also entering a new era with greater access to bariatric surgery and approval of anti-obesity medications (glucagon-like peptide-1 analogues) that produce substantial weight loss for many people, but there are concerns about disproportionate loss of lean mass and nutritional deficiencies. RECENT FINDINGS No matter the approach used to achieve major weight loss, careful attention to nutritional considerations is necessary. Here, we examine the recent findings regarding the importance of adequate protein to maintain lean mass, the rationale and evidence supporting low-carbohydrate and ketogenic dietary patterns, and the potential benefits of including exercise training in the context of major weight loss. While losing and sustaining weight loss has proven challenging, we are optimistic that application of emerging nutrition science, particularly personalized well-formulated low-carbohydrate dietary patterns that contain adequate protein (1.2 to 2.0 g per kilogram reference weight) and achieve the beneficial metabolic state of euketonemia (circulating ketones 0.5 to 5 mM), is a promising path for many individuals with excess adiposity.
Collapse
Affiliation(s)
- Jeff S Volek
- Department of Human Sciences, The Ohio State University, 305 Annie & John Glenn Ave, Columbus, OH, 43210, USA.
| | - Madison L Kackley
- Department of Human Sciences, The Ohio State University, 305 Annie & John Glenn Ave, Columbus, OH, 43210, USA
| | - Alex Buga
- Department of Human Sciences, The Ohio State University, 305 Annie & John Glenn Ave, Columbus, OH, 43210, USA
| |
Collapse
|
2
|
Bartholomew CL, Martins C, Gower B. Association between insulin sensitivity and lean mass loss during weight loss. Obesity (Silver Spring) 2024; 32:1156-1162. [PMID: 38803306 PMCID: PMC11141400 DOI: 10.1002/oby.24022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 02/21/2024] [Accepted: 02/22/2024] [Indexed: 05/29/2024]
Abstract
OBJECTIVE The study objective was to assess the relationship between insulin sensitivity and changes in total lean mass (LM) and appendicular LM (ALM) during weight loss. METHODS Individuals were randomly assigned to either a standard or a moderately reduced carbohydrate diet for 16 weeks. Body composition was assessed using dual-energy x-ray absorptiometry and insulin sensitivity index (SI) using an intravenous glucose tolerance test. Multiple linear regression was used to determine whether baseline SI was predictive of changes in total LM and ALM. RESULTS Participants (n = 57; baseline BMI 32.1 ± 3.8 kg/m2) lost an average of 6.8 ± 3.2 kg of body weight (p < 0.001), with 1.5 ± 2.6 kg coming from LM (p < 0.05) and 0.5 ± 0.73 kg from ALM (p < 0.05). Multiple regression analysis demonstrated that SI was inversely associated with changes in total LM (kilograms; β = 0.481, p < 0.001), after adjusting for baseline LM, fat mass, acute insulin response to glucose, and weight loss. Similar results were seen when assessing ALM loss (β = 0.359, p < 0.05). CONCLUSIONS Identifying individuals with low insulin sensitivity prior to weight loss interventions may allow for a personalized approach aiming at minimizing LM loss.
Collapse
Affiliation(s)
- Ciera L Bartholomew
- Department of Nutrition Sciences, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
| | - Catia Martins
- Department of Nutrition Sciences, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
| | - Barbara Gower
- Department of Nutrition Sciences, University of Alabama at Birmingham (UAB), Birmingham, Alabama, USA
| |
Collapse
|
3
|
Wong JMW, Ludwig DS, Allison DB, Baidwan N, Bielak L, Chiu CY, Dickinson SL, Golzarri-Arroyo L, Heymsfield SB, Holmes L, Jansen LT, Lesperance D, Mehta T, Sandman M, Steltz SK, Wong WW, Yu S, Ebbeling CB. Design and conduct of a randomized controlled feeding trial in a residential setting with mitigation for COVID-19. Contemp Clin Trials 2024; 140:107490. [PMID: 38458559 DOI: 10.1016/j.cct.2024.107490] [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: 12/19/2023] [Revised: 02/26/2024] [Accepted: 03/05/2024] [Indexed: 03/10/2024]
Abstract
BACKGROUND Evaluating effects of different macronutrient diets in randomized trials requires well defined infrastructure and rigorous methods to ensure intervention fidelity and adherence. METHODS This controlled feeding study comprised two phases. During a Run-in phase (14-15 weeks), study participants (18-50 years, BMI, ≥27 kg/m2) consumed a very-low-carbohydrate (VLC) diet, with home delivery of prepared meals, at an energy level to promote 15 ± 3% weight loss. During a Residential phase (13 weeks), participants resided at a conference center. They received a eucaloric VLC diet for three weeks and then were randomized to isocaloric test diets for 10 weeks: VLC (5% energy from carbohydrate, 77% from fat), high-carbohydrate (HC)-Starch (57%, 25%; including 20% energy from refined grains), or HC-Sugar (57%, 25%; including 20% sugar). Outcomes included measures of body composition and energy expenditure, chronic disease risk factors, and variables pertaining to physiological mechanisms. Six cores provided infrastructure for implementing standardized protocols: Recruitment, Diet and Meal Production, Participant Support, Assessments, Regulatory Affairs and Data Management, and Statistics. The first participants were enrolled in May 2018. Participants residing at the conference center at the start of the COVID-19 pandemic completed the study, with each core implementing mitigation plans. RESULTS Before early shutdown, 77 participants were randomized, and 70 completed the trial (65% of planned completion). Process measures indicated integrity to protocols for weighing menu items, within narrow tolerance limits, and participant adherence, assessed by direct observation and continuous glucose monitoring. CONCLUSION Available data will inform future research, albeit with less statistical power than originally planned.
Collapse
Affiliation(s)
- Julia M W Wong
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, United States of America; Harvard Medical School, Boston, MA, United States of America
| | - David S Ludwig
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, United States of America; Harvard Medical School, Boston, MA, United States of America
| | - David B Allison
- Indiana University School of Public Health, Bloomington, IN, United States of America
| | - Navneet Baidwan
- Department of Health Services Administration, School of Health Professions, University of Alabama at Birmingham, United States of America
| | - Lisa Bielak
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, United States of America
| | - Chia-Ying Chiu
- Division of Pulmonary, Allergy, and Acute Critical Care, Department of Medicine, University of Alabama at Birmingham, United States of America
| | - Stephanie L Dickinson
- Indiana University School of Public Health, Bloomington, IN, United States of America
| | | | - Steven B Heymsfield
- Metabolism & Body Composition Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, United States of America
| | - Lauren Holmes
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, United States of America
| | - Lisa T Jansen
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, United States of America; Harvard Medical School, Boston, MA, United States of America
| | - Donna Lesperance
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, United States of America
| | - Tapan Mehta
- Department of Health Services Administration, School of Health Professions, University of Alabama at Birmingham, United States of America
| | - Megan Sandman
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, United States of America
| | - Sarah K Steltz
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, United States of America
| | - William W Wong
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, United States of America
| | - Shui Yu
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, United States of America
| | - Cara B Ebbeling
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, United States of America; Harvard Medical School, Boston, MA, United States of America.
| |
Collapse
|
4
|
Soto-Mota A, Jansen LT, Norwitz NG, Pereira MA, Ebbeling CB, Ludwig DS. Physiologic Adaptation to Macronutrient Change Distorts Findings from Short Dietary Trials: Reanalysis of a Metabolic Ward Study. J Nutr 2024; 154:1080-1086. [PMID: 38128881 PMCID: PMC11347797 DOI: 10.1016/j.tjnut.2023.12.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/06/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023] Open
Abstract
An influential 2-wk cross-over feeding trial without a washout period purported to show advantages of a low-fat diet (LFD) compared with a low-carbohydrate diet (LCD) for weight control. In contrast to several other macronutrient trials, the diet order effect was originally reported as not significant. In light of a new analysis by the original investigative group identifying an order effect, we aimed to examine, in a reanalysis of publicly available data (16 of 20 original participants; 7 female; mean BMI, 27.8 kg/m2), the validity of the original results and the claims that trial data oppose the carbohydrate-insulin model of obesity (CIM). We found that energy intake on the LCD was much lower when this diet was consumed first compared with second (a difference of -1164 kcal/d, P = 3.6 × 10-13); the opposite pattern was observed for the LFD (924 kcal/d, P = 2.0 × 10-16). This carry-over effect was significant (P interaction = 0.0004) whereas the net dietary effect was not (P = 0.4). Likewise, the between-arm difference (LCD - LFD) was -320 kcal/d in the first period and +1771 kcal/d in the second. Body fat decreased with consumption of the LCD first and increased with consumption of this diet second (-0.69 ± 0.33 compared with 0.57 ± 0.32 kg, P = 0.007). LCD-first participants had higher β-hydroxybutyrate levels while consuming the LCD and lower respiratory quotients while consuming LFD when compared with LFD-first participants on their respective diets. Change in insulin secretion as assessed by C-peptide in the first diet period predicted higher energy intake and less fat loss in the second period. These findings, which tend to support rather than oppose the CIM, suggest that differential (unequal) carry-over effects and short duration, with no washout period, preclude causal inferences regarding chronic macronutrient effects from this trial.
Collapse
Affiliation(s)
- Adrian Soto-Mota
- Metabolic Diseases Research Unit. National Institute of Medical Sciences and Nutrition Salvador Zubiran. Mexico City, Mexico; Tecnologico de Monterrey. School of Medicine. Mexico City, Mexico
| | - Lisa T Jansen
- Department of Dietetics & Nutrition, University of Arkansas for Medical Sciences, Little Rock, AR, United States; Arkansas Children's Nutrition Center, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | | | - Mark A Pereira
- Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, MN, United States
| | - Cara B Ebbeling
- Harvard Medical School, Boston, MA, United States; New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Boston MA, United States
| | - David S Ludwig
- Harvard Medical School, Boston, MA, United States; New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Boston MA, United States; Department of Nutrition, Exercise and Sports, University of Copenhagen.
| |
Collapse
|
5
|
Soto-Mota A, Jansen LT, Norwitz NG, Pereira MA, Ebbeling CB, Ludwig DS. Reply to C M Sciarrillo et al. J Nutr 2024; 154:1061-1063. [PMID: 38316213 DOI: 10.1016/j.tjnut.2024.01.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 01/25/2024] [Accepted: 01/27/2024] [Indexed: 02/07/2024] Open
Affiliation(s)
- Adrian Soto-Mota
- From the Metabolic Diseases Research Unit, National Institute of Medical Sciences and Nutrition Salvador Zubiran, Mexico City, Mexico; The Tecnologico de Monterrey, School of Medicine, Mexico City, Mexico
| | - Lisa T Jansen
- The Department of Dietetics & Nutrition, University of Arkansas for Medical Sciences, Little Rock, AR, United States; The Arkansas Children's Nutrition Center, Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | | | - Mark A Pereira
- The Division of Epidemiology and Community Health, School of Public Health, University of Minnesota, MN, United States
| | - Cara B Ebbeling
- The Harvard Medical School, Boston, MA, United States; The New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Boston MA, United States
| | - David S Ludwig
- The Harvard Medical School, Boston, MA, United States; The New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Boston MA, United States; The Department of Nutrition, Exercise and Sports, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
6
|
Milhem F, Komarnytsky S. Progression to Obesity: Variations in Patterns of Metabolic Fluxes, Fat Accumulation, and Gastrointestinal Responses. Metabolites 2023; 13:1016. [PMID: 37755296 PMCID: PMC10535155 DOI: 10.3390/metabo13091016] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 09/12/2023] [Accepted: 09/13/2023] [Indexed: 09/28/2023] Open
Abstract
Obesity is a multifactorial disorder that is remarkably heterogeneous. It presents itself in a variety of phenotypes that can be metabolically unhealthy or healthy, associate with no or multiple metabolic risk factors, gain extreme body weight (super-responders), as well as resist obesity despite the obesogenic environment (non-responders). Progression to obesity is ultimately linked to the overall net energy balance and activity of different metabolic fluxes. This is particularly evident from variations in fatty acids oxidation, metabolic fluxes through the pyruvate-phosphoenolpyruvate-oxaloacetate node, and extracellular accumulation of Krebs cycle metabolites, such as citrate. Patterns of fat accumulation with a focus on visceral and ectopic adipose tissue, microbiome composition, and the immune status of the gastrointestinal tract have emerged as the most promising targets that allow personalization of obesity and warrant further investigations into the critical issue of a wider and long-term weight control. Advances in understanding the biochemistry mechanisms underlying the heterogenous obesity phenotypes are critical to the development of targeted strategies to maintain healthy weight.
Collapse
Affiliation(s)
- Fadia Milhem
- Plants for Human Health Institute, NC State University, 600 Laureate Way, Kannapolis, NC 28081, USA;
- Department of Food, Bioprocessing, and Nutrition Sciences, North Carolina State University, 400 Dan Allen Drive, Raleigh, NC 27695, USA
- Department of Nutrition, University of Petra, 317 Airport Road, Amman 11196, Jordan
| | - Slavko Komarnytsky
- Plants for Human Health Institute, NC State University, 600 Laureate Way, Kannapolis, NC 28081, USA;
- Department of Food, Bioprocessing, and Nutrition Sciences, North Carolina State University, 400 Dan Allen Drive, Raleigh, NC 27695, USA
| |
Collapse
|
7
|
Ludwig DS, Ebbeling CB, Rimm EB. Carbohydrates, Insulin Secretion, and "Precision Nutrition". Diabetes Care 2022; 45:1303-1305. [PMID: 35653600 DOI: 10.2337/dci22-0009] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 03/05/2022] [Indexed: 02/03/2023]
Affiliation(s)
- David S Ludwig
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Boston, MA.,Department of Pediatrics, Harvard Medical School, Boston, MA.,Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Cara B Ebbeling
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Boston, MA.,Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Eric B Rimm
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA
| |
Collapse
|
8
|
Wong JMW, Yu S, Ma C, Mehta T, Dickinson SL, Allison DB, Heymsfield SB, Ebbeling CB, Ludwig DS. Stimulated Insulin Secretion Predicts Changes in Body Composition Following Weight Loss in Adults with High BMI. J Nutr 2022; 152:655-662. [PMID: 34587231 DOI: 10.1093/jn/nxab315] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/02/2021] [Accepted: 08/26/2021] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND The aim of obesity treatment is to promote loss of fat relative to lean mass. However, body composition changes with calorie restriction differ among individuals. OBJECTIVES The goal of this study was to test the hypothesis that insulin secretion predicts body composition changes among young and middle-age adults with high BMI (in kg/m2) following major weight loss. METHODS Exploratory analyses were conducted with pre-randomization data from 2 large feeding trials: the Framingham, Boston, Bloomington, Birmingham, and Baylor study (FB4; n = 82, 43.9% women, BMI ≥27) and the Framingham State Food Study [(FS)2; n = 161, 69.6% women, BMI ≥25]. Participants in the 2 trials consumed calorie-restricted moderate-carbohydrate or very-low-carbohydrate diets to produce 12-18% weight loss in ∼14 wk or 10-14% in ∼10 wk, respectively. We determined insulin concentration 30 min after a 75-g oral glucose load (insulin-30) as a measure of insulin secretion and HOMA-IR as a measure of insulin resistance at baseline. Body composition was determined by DXA at baseline and post-weight loss. Associations were analyzed using general linear models with adjustment for covariates. RESULTS In FB4, higher insulin-30 was associated with a smaller decrease in fat mass (0.441 kg per 100 μIU/mL increment in baseline insulin-30; P = 0.005; -1.20-kg mean difference between the first compared with the fifth group of insulin-30) and a larger decrease in lean mass (-0.465 kg per 100 μIU/mL; P = 0.004; 1.27-kg difference). Participants with higher insulin-30 lost a smaller proportion of weight loss as fat (-3.37% per 100 μIU/mL; P = 0.003; 9.20% difference). Greater HOMA-IR was also significantly associated with adverse body composition changes. Results from (FS)2 were qualitatively similar but of a smaller magnitude. CONCLUSIONS Baseline insulin dynamics predict substantial individual differences in body composition following weight loss. These findings may inform understanding of the pathophysiological basis for weight regain and the design of more effective obesity treatment. Registered at clinicaltrials.gov as NCT03394664 and NCT02068885.
Collapse
Affiliation(s)
- Julia M W Wong
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Shui Yu
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Boston, MA, USA
| | - Clement Ma
- Harvard Medical School, Boston, MA, USA.,Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA
| | - Tapan Mehta
- Department of Health Services Administration, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - David B Allison
- Indiana University School of Public Health-Bloomington, Bloomington, IN, USA
| | - Steven B Heymsfield
- Metabolism & Body Composition Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Cara B Ebbeling
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - David S Ludwig
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| |
Collapse
|
9
|
Bikman BT, Shimy KJ, Apovian CM, Yu S, Saito ER, Walton CM, Ebbeling CB, Ludwig DS. A high-carbohydrate diet lowers the rate of adipose tissue mitochondrial respiration. Eur J Clin Nutr 2022; 76:1339-1342. [PMID: 35177807 PMCID: PMC9381644 DOI: 10.1038/s41430-022-01097-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 02/01/2022] [Accepted: 02/04/2022] [Indexed: 11/13/2022]
Abstract
Adipocyte mitochondrial respiration may influence metabolic fuel partitioning into oxidation versus storage, with implications for whole-body energy expenditure. Although insulin has been shown to influence mitochondrial respiration, the effects of dietary macronutrient composition have not been well characterized. The aim of this exploratory study was to test the hypothesis that a high-carbohydrate diet lowers the oxygen flux of adipocyte mitochondria ex vivo. Among participants in a randomized-controlled weight-loss maintenance feeding trial, those consuming a high-carbohydrate diet (60% carbohydrate as a proportion of total energy, n = 10) had lower rates of maximal adipose tissue mitochondrial respiration than those consuming a moderate-carbohydrate diet (40%, n = 8, p = 0.039) or a low-carbohydrate diet (20%, n = 9, p = 0.005) after 10 to 15 weeks. This preliminary finding may provide a mechanism for postulated calorie-independent effects of dietary composition on energy expenditure and fat deposition, potentially through the actions of insulin on fuel partitioning.
Collapse
Affiliation(s)
- Benjamin T Bikman
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT, USA.
| | - Kim J Shimy
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA, USA.,Division of Endocrinology, Children's National Hospital, Washington, DC, USA
| | - Caroline M Apovian
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital and Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - S Yu
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Erin R Saito
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT, USA
| | - Chase M Walton
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT, USA
| | - Cara B Ebbeling
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - David S Ludwig
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital and Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
10
|
Ludwig DS, Wong JMW, Yu S, Ma C, Mehta T, Dickinson SL, Allison DB, Heymsfield SB, Ebbeling CB. Reply to DA Booth. J Nutr 2022; 152:641-642. [PMID: 35137123 DOI: 10.1093/jn/nxab409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 11/26/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- David S Ludwig
- From the New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Boston, MA, USA.,Harvard Medical School, Boston MA, USA
| | - Julia M W Wong
- From the New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Boston, MA, USA.,Harvard Medical School, Boston MA, USA
| | - Shui Yu
- From the New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Boston, MA, USA
| | - Clement Ma
- Harvard Medical School, Boston MA, USA.,Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston MA, USA.,Centre for Addiction and Mental Health, Toronto, ON, Canada
| | - Tapan Mehta
- Department Health Services Administration, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - David B Allison
- Indiana University School of Public Health-Bloomington, Bloomington, IN, USA
| | - Steven B Heymsfield
- Metabolism & Body Composition Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - Cara B Ebbeling
- From the New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Boston, MA, USA.,Harvard Medical School, Boston MA, USA
| |
Collapse
|
11
|
Ludwig DS, Aronne LJ, Astrup A, de Cabo R, Cantley LC, Friedman MI, Heymsfield SB, Johnson JD, King JC, Krauss RM, Lieberman DE, Taubes G, Volek JS, Westman EC, Willett WC, Yancy WS, Ebbeling CB. The carbohydrate-insulin model: a physiological perspective on the obesity pandemic. Am J Clin Nutr 2021; 114:1873-1885. [PMID: 34515299 PMCID: PMC8634575 DOI: 10.1093/ajcn/nqab270] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 07/26/2021] [Indexed: 12/29/2022] Open
Abstract
According to a commonly held view, the obesity pandemic is caused by overconsumption of modern, highly palatable, energy-dense processed foods, exacerbated by a sedentary lifestyle. However, obesity rates remain at historic highs, despite a persistent focus on eating less and moving more, as guided by the energy balance model (EBM). This public health failure may arise from a fundamental limitation of the EBM itself. Conceptualizing obesity as a disorder of energy balance restates a principle of physics without considering the biological mechanisms that promote weight gain. An alternative paradigm, the carbohydrate-insulin model (CIM), proposes a reversal of causal direction. According to the CIM, increasing fat deposition in the body-resulting from the hormonal responses to a high-glycemic-load diet-drives positive energy balance. The CIM provides a conceptual framework with testable hypotheses for how various modifiable factors influence energy balance and fat storage. Rigorous research is needed to compare the validity of these 2 models, which have substantially different implications for obesity management, and to generate new models that best encompass the evidence.
Collapse
Affiliation(s)
- David S Ludwig
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Louis J Aronne
- Comprehensive Weight Control Center, Weill Cornell Medicine, New York, NY, USA
| | - Arne Astrup
- Department of Nutrition, Exercise, and Sports, University of Copenhagen, Copenhagen, Denmark
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, USA
| | - Lewis C Cantley
- Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Mark I Friedman
- Monell Chemical Senses Center, Philadelphia, PA, USA
- Nutrition Science Initiative, San Diego, CA, USA
| | - Steven B Heymsfield
- Metabolism & Body Composition Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - James D Johnson
- Diabetes Research Group, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
- Institute for Personalized Therapeutic Nutrition, Vancouver, British Columbia, Canada
| | - Janet C King
- Department of Nutritional Sciences & Toxicology, University of California Berkeley, Berkeley, CA, USA
| | - Ronald M Krauss
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA
- Department of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Daniel E Lieberman
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Gary Taubes
- Nutrition Science Initiative, San Diego, CA, USA
| | - Jeff S Volek
- Department of Human Sciences, Ohio State University, Columbus, OH, USA
| | - Eric C Westman
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Walter C Willett
- Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - William S Yancy
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Cara B Ebbeling
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
12
|
Landry MJ, Crimarco A, Gardner CD. Benefits of Low Carbohydrate Diets: a Settled Question or Still Controversial? Curr Obes Rep 2021; 10:409-422. [PMID: 34297345 PMCID: PMC9621749 DOI: 10.1007/s13679-021-00451-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/31/2021] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW The purpose of this review was to provide an update on the available data on the benefits of low-carbohydrate (low-carb) diets for weight management and type 2 diabetes (T2DM) and determine if low-carb diets were a settled question or still controversial. RECENT FINDINGS Most of the recent published literature in this area consists of reviews of past trials, with a relatively smaller number of recent trials. Low-carb is most commonly compared to low-fat, with problematically inconsistent definitions of both. There are numerous challenges in trying to draw clear conclusions about efficacy and effectiveness. Short-term vs. long-term effects can differ, which is likely impacted by adherence. Adherence is very different between metabolic chamber or feeding studies vs. free-living. Body weight alone is a crude measure that fails to capture potentially important differences in lean-mass, fat-mass, and body water. Benefits for glycemic control need to be balanced with impacts on non-glycemic outcomes such as LDL-cholesterol, the microbiome, and inflammation. It is important to differentiate between low-carb and very-low carbohydrate diets (VLCD). To date no large-scale long-term clinical trials have been conducted testing whether low-carb diets can prevent T2DM. Many issues regarding benefits and risks of low-carb diets remain controversial or unresolved, particularly for VLCD. Some of the recent, better studies highlighted in this review suggest strategies for resolving these controversies.
Collapse
Affiliation(s)
- Matthew J Landry
- Stanford Prevention Research Center, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Anthony Crimarco
- Stanford Prevention Research Center, School of Medicine, Stanford University, Palo Alto, CA, USA
| | - Christopher D Gardner
- Stanford Prevention Research Center, School of Medicine, Stanford University, Palo Alto, CA, USA.
| |
Collapse
|
13
|
Holsen LM, Hoge WS, Lennerz BS, Cerit H, Hye T, Moondra P, Goldstein JM, Ebbeling CB, Ludwig DS. Diets Varying in Carbohydrate Content Differentially Alter Brain Activity in Homeostatic and Reward Regions in Adults. J Nutr 2021; 151:2465-2476. [PMID: 33852013 PMCID: PMC8349124 DOI: 10.1093/jn/nxab090] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 02/18/2021] [Accepted: 03/11/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Obesity has one of the highest refractory rates of all chronic diseases, in part because weight loss induced by calorie restriction, the first-line treatment for obesity, elicits biological adaptations that promote weight regain. Although acute feeding trials suggest a role for macronutrient composition in modifying brain activity related to hunger and satiety, relevance of these findings to weight-loss maintenance has not been studied. OBJECTIVES We investigated effects of weight-loss maintenance diets varying in macronutrient content on regional cerebral blood flow (rCBF) in brain regions involved in hunger and reward. METHODS In conjunction with a randomized controlled feeding trial, we investigated the effects of weight-loss maintenance diets varying in carbohydrate content [high, 60% of total energy: n = 20; 6 men/14 women; mean age: 32.5 y; mean BMI (in kg/m 2): 27.4; moderate, 40% of total energy: n = 22; 10 men/12 women; mean age: 32.5 y; mean BMI: 29.0; low, 20% of total energy: n = 28; 12 men/16 women; mean age: 33.2 y; mean BMI: 27.7] on rCBF in brain regions involved in hunger and reward preprandial and 4 h postprandial after 14-20 wk on the diets. The primary outcome was rCBF in the nucleus accumbens (NAcc) at 4 h postprandial; the secondary outcome was preprandial rCBF in the hypothalamus. RESULTS Consistent with a priori hypothesis, at 4 h postprandial, NAcc rCBF was 43% higher in adults assigned to the high- compared with low-carbohydrate diet {P[family-wise error (FWE)-corrected] < 0.05}. Preprandial hypothalamus rCBF was 41% higher on high-carbohydrate diet [P(FWE-corrected) < 0.001]. Exploratory analyses revealed that elevated rCBF on high-carbohydrate diet was not specific to prandial state: preprandial NAcc rCBF [P(FWE-corrected) < 0.001] and 4 h postprandial rCBF in hypothalamus [P(FWE-corrected) < 0.001]. Insulin secretion predicted differential postprandial activation of the NAcc by diet. CONCLUSIONS We report significant differences in rCBF in adults assigned to diets varying in carbohydrate content for several months, which appear to be partially associated with insulin secretion. These findings suggest that chronic intake of a high-carbohydrate diet may affect brain reward and homeostatic activity in ways that could impede weight-loss maintenance. This trial was registered at clinicaltrials.gov as NCT02300857.
Collapse
Affiliation(s)
- Laura M Holsen
- Division of Women's Health and Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Department of Psychiatry, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - W Scott Hoge
- Harvard Medical School, Boston, MA, USA
- Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | - Belinda S Lennerz
- Harvard Medical School, Boston, MA, USA
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, Boston, MA, USA
| | - Hilâl Cerit
- Division of Women's Health and Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Taryn Hye
- Division of Women's Health and Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Priyanka Moondra
- Division of Women's Health and Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Jill M Goldstein
- Division of Women's Health and Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Psychiatry and Department of Obstetrics and Gynecology, Massachusetts General Hospital, Boston, MA, USA
| | - Cara B Ebbeling
- Harvard Medical School, Boston, MA, USA
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, Boston, MA, USA
| | - David S Ludwig
- Harvard Medical School, Boston, MA, USA
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, Boston, MA, USA
| |
Collapse
|
14
|
Ludwig DS, Dickinson SL, Henschel B, Ebbeling CB, Allison DB. Do Lower-Carbohydrate Diets Increase Total Energy Expenditure? An Updated and Reanalyzed Meta-Analysis of 29 Controlled-Feeding Studies. J Nutr 2020; 151:482-490. [PMID: 33274750 PMCID: PMC7948201 DOI: 10.1093/jn/nxaa350] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 09/29/2020] [Accepted: 10/12/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND The effect of macronutrient composition on total energy expenditure (TEE) remains controversial, with divergent findings among studies. One source of heterogeneity may be study duration, as physiological adaptation to lower carbohydrate intake may require 2 to 3 wk. OBJECTIVE We tested the hypothesis that the effects of carbohydrate [expressed as % of energy intake (EI)] on TEE vary with time. METHODS The sample included trials from a previous meta-analysis and new trials identified in a PubMed search through 9 March 2020 comparing lower- and higher-carbohydrate diets, controlled for EI or body weight. Three reviewers independently extracted data and reconciled discrepancies. Effects on TEE were pooled using inverse-variance-weighted meta-analysis, with between-study heterogeneity assessed using the I2 statistic. Meta-regression was used to quantify the influence of study duration, dichotomized at 2.5 wk. RESULTS The 29 trials ranged in duration from 1 to 140 d (median: 4 d) and included 617 participants. Difference in carbohydrate between intervention arms ranged from 8% to 77% EI (median: 30%). Compared with reported findings in the prior analysis (I2 = 32.2%), we found greater heterogeneity (I2 = 90.9% in the reanalysis, 81.6% in the updated analysis). Study duration modified the diet effect on TEE (P < 0.001). Among 23 shorter trials, TEE was reduced on lower-carbohydrate diets (-50.0 kcal/d; 95% CI: -77.4, -22.6 kcal/d) with substantial heterogeneity (I2 = 69.8). Among 6 longer trials, TEE was increased on low-carbohydrate diets (135.4 kcal/d; 95% CI: 72.0, 198.7 kcal/d) with low heterogeneity (I2 = 26.4). Expressed per 10% decrease in carbohydrate as %EI, the TEE effects in shorter and longer trials were -14.5 kcal/d and 50.4 kcal/d, respectively. Findings were materially unchanged in sensitivity analyses. CONCLUSIONS Lower-carbohydrate diets transiently reduce TEE, with a larger increase after ∼2.5 wk. These findings highlight the importance of longer trials to understand chronic macronutrient effects and suggest a mechanism whereby lower-carbohydrate diets may facilitate weight loss.
Collapse
Affiliation(s)
| | | | - Beate Henschel
- Indiana University School of Public Health–Bloomington, Bloomington, IN, USA
| | - Cara B Ebbeling
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - David B Allison
- Indiana University School of Public Health–Bloomington, Bloomington, IN, USA
| |
Collapse
|
15
|
Gonzalez-Gil AM, Elizondo-Montemayor L. The Role of Exercise in the Interplay between Myokines, Hepatokines, Osteokines, Adipokines, and Modulation of Inflammation for Energy Substrate Redistribution and Fat Mass Loss: A Review. Nutrients 2020; 12:E1899. [PMID: 32604889 PMCID: PMC7353393 DOI: 10.3390/nu12061899] [Citation(s) in RCA: 127] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/18/2020] [Accepted: 06/18/2020] [Indexed: 12/17/2022] Open
Abstract
Exercise is an effective strategy for preventing and treating obesity and its related cardiometabolic disorders, resulting in significant loss of body fat mass, white adipose tissue browning, redistribution of energy substrates, optimization of global energy expenditure, enhancement of hypothalamic circuits that control appetite-satiety and energy expenditure, and decreased systemic inflammation and insulin resistance. Novel exercise-inducible soluble factors, including myokines, hepatokines, and osteokines, and immune cytokines and adipokines are hypothesized to play an important role in the body's response to exercise. To our knowledge, no review has provided a comprehensive integrative overview of these novel molecular players and the mechanisms involved in the redistribution of metabolic fuel during and after exercise, the loss of weight and fat mass, and reduced inflammation. In this review, we explain the potential role of these exercise-inducible factors, namely myokines, such as irisin, IL-6, IL-15, METRNL, BAIBA, and myostatin, and hepatokines, in particular selenoprotein P, fetuin A, FGF21, ANGPTL4, and follistatin. We also describe the function of osteokines, specifically osteocalcin, and of adipokines such as leptin, adiponectin, and resistin. We also emphasize an integrative overview of the pleiotropic mechanisms, the metabolic pathways, and the inter-organ crosstalk involved in energy expenditure, fat mass loss, reduced inflammation, and healthy weight induced by exercise.
Collapse
Affiliation(s)
- Adrian M. Gonzalez-Gil
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey N.L. 64710, Mexico;
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition and Obesity, Ave. Morones Prieto 300, Monterrey N.L. 64710, Mexico
| | - Leticia Elizondo-Montemayor
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, Monterrey N.L. 64710, Mexico;
- Tecnologico de Monterrey, Center for Research in Clinical Nutrition and Obesity, Ave. Morones Prieto 300, Monterrey N.L. 64710, Mexico
- Tecnologico de Monterrey, Cardiovascular and Metabolomics Research Group, Hospital Zambrano Hellion, San Pedro Garza Garcia P.C. 66278, Mexico
| |
Collapse
|
16
|
Shimy KJ, Feldman HA, Klein GL, Bielak L, Ebbeling CB, Ludwig DS. Effects of Dietary Carbohydrate Content on Circulating Metabolic Fuel Availability in the Postprandial State. J Endocr Soc 2020; 4:bvaa062. [PMID: 32666008 PMCID: PMC7326475 DOI: 10.1210/jendso/bvaa062] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 05/20/2020] [Indexed: 12/18/2022] Open
Abstract
Context According to the carbohydrate-insulin model of obesity, an elevated insulin-to-glucagon ratio in response to a high-carbohydrate diet directs metabolic fuels toward storage, resulting in lower circulating energy. Objective To determine differences in total circulating energy post-meal related to dietary carbohydrate. Design Ancillary study within the Framingham State Food Study. Setting University community. Participants 29 adults (aged 20 to 65 years) with overweight or obesity (body mass index ≥25 kg/m2) Intervention After achieving 10% to 14% weight loss on a run-in diet, participants were randomized to weight-loss-maintenance test diets varying in carbohydrate content (high-carbohydrate, 60% of total energy, n = 11; moderate-carbohydrate, 40%, n = 8; low-carbohydrate, 20%, n = 10) and controlled for protein (20%). During 24-hour metabolic ward admissions between 10 and 15 weeks on the test diets, metabolic fuels and hormones were measured. Main Outcome Measure Energy availability (EA) based on energy content of blood glucose, beta-hydroxybutyrate, and free fatty acids, in the late postprandial period (180 to 300 minutes). Insulin at 30 minutes into the test meal (Meal Insulin-30) was measured as an effect modifier. Results Insulin-to-glucagon ratio was 7-fold higher in participants on the high- vs low-carbohydrate diet (2.5 and 0.36, respectively). Late postprandial EA was 0.58 kcal/L lower on the high- vs low-carbohydrate diet (P < 0.0001), primarily related to suppression of free fatty acids. Early postprandial EA (30 to 180 minutes) declined fastest in the high-carbohydrate group, and Meal Insulin-30 modified this diet effect. Conclusions During weight-loss maintenance on a high-carbohydrate diet, late postprandial EA is reduced, consistent with the carbohydrate-insulin model.
Collapse
Affiliation(s)
- Kim J Shimy
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Boston, Massachusetts.,Division of Endocrinology, Children's National Medical Center, Washington, DC
| | - Henry A Feldman
- Institutional Centers for Clinical and Translational Research, Boston Children's Hospital, Boston, Massachusetts
| | - Gloria L Klein
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Boston, Massachusetts
| | - Lisa Bielak
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Boston, Massachusetts
| | - Cara B Ebbeling
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| | - David S Ludwig
- New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Boston, Massachusetts.,Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
17
|
Most J, Redman LM. Impact of calorie restriction on energy metabolism in humans. Exp Gerontol 2020; 133:110875. [DOI: 10.1016/j.exger.2020.110875] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/06/2020] [Accepted: 02/09/2020] [Indexed: 11/28/2022]
|
18
|
Hu S, Wang L, Togo J, Yang D, Xu Y, Wu Y, Douglas A, Speakman JR. The carbohydrate-insulin model does not explain the impact of varying dietary macronutrients on the body weight and adiposity of mice. Mol Metab 2019; 32:27-43. [PMID: 32029228 PMCID: PMC6938849 DOI: 10.1016/j.molmet.2019.11.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Accepted: 11/08/2019] [Indexed: 12/24/2022] Open
Abstract
Objectives The carbohydrate-insulin model (CIM) predicts that increases in fasting and post-prandial insulin in response to dietary carbohydrates stimulate energy intake and lower energy expenditures, leading to positive energy balance and weight gain. The objective of the present study was to directly test the CIM's predictions using C57BL/6 mice. Methods Diets were designed by altering dietary carbohydrates with either fixed protein or fat content and were fed to C57BL/6 mice acutely or chronically for 12 weeks. The body weight, body composition, food intake, and energy expenditures of the mice were measured. Their fasting and post-prandial glucose and insulin levels were also measured. RNA-seq was performed on RNA from the hypothalamus and subcutaneous white adipose tissue. Pathway analysis was conducted using IPA. Results Only the post-prandial insulin and fasting glucose levels followed the CIM's predictions. The lipolysis and leptin signaling pathways in the sWAT were inhibited in relation to the elevated fasting insulin, supporting the CIM's predicted impact of high insulin. However, because higher fasting insulin was unrelated to carbohydrate intake, the overall pattern did not support the model. Moreover, the hypothalamic hunger pathways were inhibited in relation to the increased fasting insulin, and the energy intake was not increased. The browning pathway in the sWAT was inhibited at higher insulin levels, but the daily energy expenditure was not altered. Conclusions Two of the predictions were partially supported (and hence also partially not supported) and the other three predictions were not supported. We conclude that the CIM does not explain the impact of dietary macronutrients on adiposity in mice. Higher fasting insulin related to inhibited lipolysis and leptin pathways in sWAT, supporting CIM. Higher fasting insulin related to inhibited hypothalamic hunger pathway, contrasting CIM. Fasting insulin decreased with higher dietary carbohydrate, overall contrasting CIM. Higher dietary carbohydrate did not lead to greater EI/adiposity, or lowered EE.
Collapse
Affiliation(s)
- Sumei Hu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Lu Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China; University of Chinese Academy of Sciences, Shijingshan District, Beijing, 100049, PR China; Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, AB24 2TZ, Scotland, UK
| | - Jacques Togo
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China; University of Chinese Academy of Sciences, Shijingshan District, Beijing, 100049, PR China
| | - Dengbao Yang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Yanchao Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China
| | - Yingga Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China; University of Chinese Academy of Sciences, Shijingshan District, Beijing, 100049, PR China; Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, AB24 2TZ, Scotland, UK
| | - Alex Douglas
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, AB24 2TZ, Scotland, UK
| | - John R Speakman
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, PR China; Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, AB24 2TZ, Scotland, UK; CAS Center for Excellence in Animal Evolution and Genetics (CCEAEG), Kunming, PR China.
| |
Collapse
|
19
|
Astley CM, Todd JN, Salem RM, Vedantam S, Ebbeling CB, Huang PL, Ludwig DS, Hirschhorn JN, Florez JC. Genetic Evidence That Carbohydrate-Stimulated Insulin Secretion Leads to Obesity. Clin Chem 2019; 64:192-200. [PMID: 29295838 DOI: 10.1373/clinchem.2017.280727] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 10/17/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND A fundamental precept of the carbohydrate-insulin model of obesity is that insulin secretion drives weight gain. However, fasting hyperinsulinemia can also be driven by obesity-induced insulin resistance. We used genetic variation to isolate and estimate the potentially causal effect of insulin secretion on body weight. METHODS Genetic instruments of variation of insulin secretion [assessed as insulin concentration 30 min after oral glucose (insulin-30)] were used to estimate the causal relationship between increased insulin secretion and body mass index (BMI), using bidirectional Mendelian randomization analysis of genome-wide association studies. Data sources included summary results from the largest published metaanalyses of predominantly European ancestry for insulin secretion (n = 26037) and BMI (n = 322154), as well as individual-level data from the UK Biobank (n = 138541). Data from the Cardiology and Metabolic Patient Cohort study at Massachusetts General Hospital (n = 1675) were used to validate genetic associations with insulin secretion and to test the observational association of insulin secretion and BMI. RESULTS Higher genetically determined insulin-30 was strongly associated with higher BMI (β = 0.098, P = 2.2 × 10-21), consistent with a causal role in obesity. Similar positive associations were noted in sensitivity analyses using other genetic variants as instrumental variables. By contrast, higher genetically determined BMI was not associated with insulin-30. CONCLUSIONS Mendelian randomization analyses provide evidence for a causal relationship of glucose-stimulated insulin secretion on body weight, consistent with the carbohydrate-insulin model of obesity.
Collapse
Affiliation(s)
- Christina M Astley
- Department of Medicine, Division of Endocrinology, Boston Children's Hospital, Boston, MA.,Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA.,Programs in Metabolism and Medical & Population Genetics, Broad Institute, Cambridge, MA
| | - Jennifer N Todd
- Department of Medicine, Division of Endocrinology, Boston Children's Hospital, Boston, MA.,Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA.,Programs in Metabolism and Medical & Population Genetics, Broad Institute, Cambridge, MA.,Diabetes Unit and Center for Genomic Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - Rany M Salem
- Department of Family Medicine and Public Health, University of California, San Diego, CA
| | - Sailaja Vedantam
- Department of Medicine, Division of Endocrinology, Boston Children's Hospital, Boston, MA.,Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA
| | - Cara B Ebbeling
- Department of Medicine, Division of Endocrinology, Boston Children's Hospital, Boston, MA.,Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA
| | - Paul L Huang
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA.,Cardiology Division, Department of Medicine, Massachusetts General Hospital, Boston, MA
| | - David S Ludwig
- Department of Medicine, Division of Endocrinology, Boston Children's Hospital, Boston, MA; .,Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA
| | - Joel N Hirschhorn
- Department of Medicine, Division of Endocrinology, Boston Children's Hospital, Boston, MA; .,Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA.,Programs in Metabolism and Medical & Population Genetics, Broad Institute, Cambridge, MA
| | - Jose C Florez
- Departments of Medicine and Pediatrics, Harvard Medical School, Boston, MA; .,Programs in Metabolism and Medical & Population Genetics, Broad Institute, Cambridge, MA.,Diabetes Unit and Center for Genomic Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
20
|
Hall KD, Guo J, Chen KY, Leibel RL, Reitman ML, Rosenbaum M, Smith SR, Ravussin E. Methodologic considerations for measuring energy expenditure differences between diets varying in carbohydrate using the doubly labeled water method. Am J Clin Nutr 2019; 109:1328-1334. [PMID: 31028699 PMCID: PMC6499509 DOI: 10.1093/ajcn/nqy390] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 12/20/2018] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Low-carbohydrate diets have been reported to significantly increase human energy expenditure when measured using doubly labeled water (DLW) but not by respiratory chambers. Although DLW may reveal true physiological differences undetected by respiratory chambers, an alternative possibility is that the expenditure differences resulted from failure to correctly estimate the respiratory quotient (RQ) used in the DLW calculations. OBJECTIVE To examine energy expenditure differences between isocaloric diets varying widely in carbohydrate and to quantitatively compare DLW data with respiratory chamber and body composition measurements within an energy balance framework. DESIGN DLW measurements were obtained during the final 2 wk of month-long baseline (BD; 50% carbohydrate, 35% fat, 15% protein) and isocaloric ketogenic diets (KD; 5% carbohydrate, 80% fat, 15% protein) in 17 men with a BMI of 25-35 kg/m2. Subjects resided 2 d/wk in respiratory chambers to measure energy expenditure (EEchamber). DLW expenditure was calculated using chamber-determined RQ either unadjusted (EEDLW) or adjusted (EEDLWΔRQ) for net energy imbalance using diet-specific coefficients. Accelerometers measured physical activity. Body composition changes were measured by dual-energy X-ray absorptiometry (DXA) which were combined with energy intake measurements to calculate energy expenditure by balance (EEbal). RESULTS After transitioning from BD to KD, neither EEchamber nor EEbal were significantly changed (∆EEchamber = 24 ± 30 kcal/d; P = 0.43 and ∆EEbal = -141 ± 118 kcal/d; P = 0.25). Similarly, physical activity (-5.1 ± 4.8%; P = 0.3) and exercise efficiency (-1.6 ± 2.4%; P = 0.52) were not significantly changed. However, EEDLW was 209 ± 83 kcal/d higher during the KD (P = 0.023) but was not significantly increased when adjusted for energy balance (EEDLWΔRQ = 139 ± 89 kcal/d; P = 0.14). After removing 2 outliers whose EEDLW were incompatible with other data, EEDLW was marginally increased during the KD by 126 ± 62 kcal/d (P = 0.063) and EEDLW∆RQ was only 46 ± 65 kcal/d higher (P = 0.49). CONCLUSIONS DLW calculations failing to account for diet-specific energy imbalance effects on RQ erroneously suggest that low-carbohydrate diets substantially increase energy expenditure. This trial was registered at clinicaltrials.gov as NCT01967563.
Collapse
Affiliation(s)
- Kevin D Hall
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | - Juen Guo
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | - Kong Y Chen
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | | | - Marc L Reitman
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD
| | | | - Steven R Smith
- The Translational Research Institute for Metabolism and Diabetes, Orlando, FL
| | - Eric Ravussin
- Pennington Biomedical Research Center, Baton Rouge, LA
| |
Collapse
|
21
|
The Metabolomic Signatures of Weight Change. Metabolites 2019; 9:metabo9040067. [PMID: 30987392 PMCID: PMC6523676 DOI: 10.3390/metabo9040067] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 03/18/2019] [Accepted: 04/03/2019] [Indexed: 12/17/2022] Open
Abstract
Obesity represents a major health concern, not just in the West but increasingly in low and middle income countries. In order to develop successful strategies for losing weight, it is essential to understand the molecular pathogenesis of weight change. A number of pathways, implicating oxidative stress but also the fundamental regulatory of insulin, have been implicated in weight gain and in the regulation of energy expenditure. In addition, a considerable body of work has highlighted the role of metabolites generated by the gut microbiome, in particular short chain fatty acids, in both processes. The current review provides a brief understanding of the mechanisms underlying the associations of weight change with changes in lipid and amino acid metabolism, energy metabolism, dietary composition and insulin dynamics, as well as the influence of the gut microbiome. The changes in metabolomic profiles and the models outlined can be used as an accurate predictor for obesity and obesity related disorders.
Collapse
|
22
|
Ebbeling CB, Feldman HA, Klein GL, Wong JMW, Bielak L, Steltz SK, Luoto PK, Wolfe RR, Wong WW, Ludwig DS. Effects of a low carbohydrate diet on energy expenditure during weight loss maintenance: randomized trial. BMJ 2018; 363:k4583. [PMID: 30429127 PMCID: PMC6233655 DOI: 10.1136/bmj.k4583] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/24/2018] [Indexed: 12/13/2022]
Abstract
OBJECTIVE To determine the effects of diets varying in carbohydrate to fat ratio on total energy expenditure. DESIGN Randomized trial. SETTING Multicenter collaboration at US two sites, August 2014 to May 2017. PARTICIPANTS 164 adults aged 18-65 years with a body mass index of 25 or more. INTERVENTIONS After 12% (within 2%) weight loss on a run-in diet, participants were randomly assigned to one of three test diets according to carbohydrate content (high, 60%, n=54; moderate, 40%, n=53; or low, 20%, n=57) for 20 weeks. Test diets were controlled for protein and were energy adjusted to maintain weight loss within 2 kg. To test for effect modification predicted by the carbohydrate-insulin model, the sample was divided into thirds of pre-weight loss insulin secretion (insulin concentration 30 minutes after oral glucose). MAIN OUTCOME MEASURES The primary outcome was total energy expenditure, measured with doubly labeled water, by intention-to-treat analysis. Per protocol analysis included participants who maintained target weight loss, potentially providing a more precise effect estimate. Secondary outcomes were resting energy expenditure, measures of physical activity, and levels of the metabolic hormones leptin and ghrelin. RESULTS Total energy expenditure differed by diet in the intention-to-treat analysis (n=162, P=0.002), with a linear trend of 52 kcal/d (95% confidence interval 23 to 82) for every 10% decrease in the contribution of carbohydrate to total energy intake (1 kcal=4.18 kJ=0.00418 MJ). Change in total energy expenditure was 91 kcal/d (95% confidence interval -29 to 210) greater in participants assigned to the moderate carbohydrate diet and 209 kcal/d (91 to 326) greater in those assigned to the low carbohydrate diet compared with the high carbohydrate diet. In the per protocol analysis (n=120, P<0.001), the respective differences were 131 kcal/d (-6 to 267) and 278 kcal/d (144 to 411). Among participants in the highest third of pre-weight loss insulin secretion, the difference between the low and high carbohydrate diet was 308 kcal/d in the intention-to-treat analysis and 478 kcal/d in the per protocol analysis (P<0.004). Ghrelin was significantly lower in participants assigned to the low carbohydrate diet compared with those assigned to the high carbohydrate diet (both analyses). Leptin was also significantly lower in participants assigned to the low carbohydrate diet (per protocol). CONCLUSIONS Consistent with the carbohydrate-insulin model, lowering dietary carbohydrate increased energy expenditure during weight loss maintenance. This metabolic effect may improve the success of obesity treatment, especially among those with high insulin secretion. TRIAL REGISTRATION ClinicalTrials.gov NCT02068885.
Collapse
Affiliation(s)
- Cara B Ebbeling
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Henry A Feldman
- Harvard Medical School, Boston, MA, USA
- Institutional Centers for Clinical and Translational Research, Boston Children's Hospital, Boston, MA, USA
| | - Gloria L Klein
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Julia M W Wong
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA, USA
| | - Lisa Bielak
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Sarah K Steltz
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Patricia K Luoto
- Department of Food and Nutrition, Framingham State University, Framingham, MA, USA
| | - Robert R Wolfe
- University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - William W Wong
- USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - David S Ludwig
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
- Harvard Medical School, Boston, MA, USA
| |
Collapse
|
23
|
Gower BA, Goss AM. The sliding set-point: how insulin and diet interact to explain the obesity epidemic (and how to fix it). Curr Opin Endocrinol Diabetes Obes 2018; 25:303-309. [PMID: 30036193 PMCID: PMC9126286 DOI: 10.1097/med.0000000000000426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
PURPOSE OF REVIEW The current approach to weight loss (intentional energy deficit) is difficult to implement and sustain, and rarely leads to successful long-term weight loss maintenance. The aim of this article is to review recent literature on the role of insulin in obesity propensity, and by extension, the effectiveness of carbohydrate restriction in facilitating weight loss, with particular attention to individual variability in patient response. RECENT FINDINGS A genetic signature for insulin secretion predisposes to elevated BMI. A genetic signature for insulin resistance is a marker for impaired fat storage, is associated with relative leanness, and predisposes to cardiometabolic disease. The largest randomized weight-loss trial ever conducted to examine insulin/diet interactions revealed no interactive effect of insulin phenotype with diet composition on body weight in the context of energy restriction. However, smaller studies revealed unique effects of carbohydrate restriction on energy partitioning that are not reflected in body weight; that is, preferential loss of total and ectopic adipose tissue. Carbohydrate-restricted diets are associated with greater adherence, and with greater total and resting energy expenditure. SUMMARY For patients with a predisposition to high insulin secretion, carbohydrate restriction may facilitate long-term reductions in body fat, perhaps by reducing hunger, maintaining energy expenditure, and promoting adherence.
Collapse
Affiliation(s)
- Barbara A Gower
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | | |
Collapse
|
24
|
Hjorth MF, Zohar Y, Hill JO, Astrup A. Personalized Dietary Management of Overweight and Obesity Based on Measures of Insulin and Glucose. Annu Rev Nutr 2018; 38:245-272. [PMID: 29856931 PMCID: PMC9105825 DOI: 10.1146/annurev-nutr-082117-051606] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
During the past several decades, numerous trials have compared various diets for the management of overweight and obesity, assuming that a single dietary strategy would be appropriate for all individuals. These studies have failed to provide strong evidence for the efficacy of any particular diet, and it is likely that different people will have different levels of success on different diets. We identified studies investigating pretreatment glycemia or insulinemia status, or both, of the individual as prognostic markers of weight loss during periods in which the composition of a participant's diet was known. Overall, research suggests that providing specific diets for weight management based on pretreatment glycemia and insulinemia statuses holds great promise for advancing personalized nutrition.
Collapse
Affiliation(s)
- Mads F Hjorth
- Department of Nutrition, Exercise, and Sports, Faculty of Sciences, University of Copenhagen, 1958 Frederiksberg C, Denmark; ,
| | | | - James O Hill
- Colorado Nutrition Obesity Research Center, University of Colorado Denver, Aurora, Colorado 80045, USA;
| | - Arne Astrup
- Department of Nutrition, Exercise, and Sports, Faculty of Sciences, University of Copenhagen, 1958 Frederiksberg C, Denmark; ,
| |
Collapse
|
25
|
Cohen CW, Fontaine KR, Arend RC, Alvarez RD, Leath III CA, Huh WK, Bevis KS, Kim KH, Straughn JM, Gower BA. A Ketogenic Diet Reduces Central Obesity and Serum Insulin in Women with Ovarian or Endometrial Cancer. J Nutr 2018; 148:1253-1260. [PMID: 30137481 PMCID: PMC8496516 DOI: 10.1093/jn/nxy119] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 05/09/2018] [Indexed: 12/11/2022] Open
Abstract
Background The glycolytic nature of cancer cells presents a potential treatment target that may be addressed by a ketogenic diet (KD). Objective We hypothesized that a KD would improve body composition and lower serum insulin and insulin-like growth factor-I (IGF-I) in women with ovarian or endometrial cancer. Methods In this randomized controlled trial, women with ovarian or endometrial cancer [age: ≥19 y; body mass index (kg/m2): ≥18.5] were randomly assigned to a KD (70:25:5 energy from fat, protein, and carbohydrate) or the American Cancer Society diet (ACS; high-fiber, low-fat). Body composition (DXA) and fasting serum insulin, IGF-I, and β-hydroxybutyrate were obtained at baseline and at 12 wk; urinary ketones were also measured throughout the intervention. We assessed differences between the diets with ANCOVA and independent t tests. We used correlation analyses to estimate associations between changes in serum analytes and body composition. Results After 12 wk, the KD (compared with ACS) group had lower adjusted total (35.3 compared with 38.0 kg, P < 0.05) and android (3.0 compared with 3.3 kg, P < 0.05) fat mass. Percentage of change in visceral fat was greater in the KD group (compared with the ACS group; -21.2% compared with -4.6%, P < 0.05). Adjusted total lean mass did not differ between the groups. The KD (compared with ACS) group had lower adjusted fasting serum insulin (7.6 compared with 11.2 µU/mL, P < 0.01). There was a significant inverse association between the changes in serum β-hydroxybutyrate and IGF-I concentrations (r = -0.57; P < 0.0001). Conclusions In women with ovarian or endometrial cancer, a KD results in selective loss of fat mass and retention of lean mass. Visceral fat mass and fasting serum insulin also are reduced by the KD, perhaps owing to enhanced insulin sensitivity. Elevated serum β-hydroxybutyrate may reflect a metabolic environment inhospitable to cancer proliferation. This trial was registered at www.clinicaltrials.gov as NCT03171506.
Collapse
Affiliation(s)
| | - Kevin R Fontaine
- Departments of Health Behavior, University of Alabama at Birmingham, Birmingham, AL
| | - Rebecca C Arend
- Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL
| | - Ronald D Alvarez
- Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL
| | - Charles A Leath III
- Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL
| | - Warner K Huh
- Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL
| | - Kerri S Bevis
- Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL
| | - Kenneth H Kim
- Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL
| | - John M Straughn
- Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, AL
| | - Barbara A Gower
- Departments of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
26
|
Page MM, Johnson JD. Mild Suppression of Hyperinsulinemia to Treat Obesity and Insulin Resistance. Trends Endocrinol Metab 2018; 29:389-399. [PMID: 29665988 DOI: 10.1016/j.tem.2018.03.018] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/23/2018] [Accepted: 03/27/2018] [Indexed: 12/14/2022]
Abstract
Insulin plays roles in lipid uptake, lipolysis, and lipogenesis, in addition to controlling blood glucose levels. Excessive circulating insulin is associated with adipose tissue expansion and obesity, yet a causal role for hyperinsulinemia in the development of mammalian obesity has proven controversial, with many researchers suggesting it as a consequence of insulin resistance. Recently, evidence that specifically reducing hyperinsulinemia can prevent and reverse obesity in animal models has been presented. Our experiments, and others in this field, question the current dogma that hyperinsulinemia is a response to obesity and/or insulin resistance. In this review, we discuss preclinical evidence in the context of the broader literature and speculate on the possibility of clinical translation of alternative approaches for treating obesity.
Collapse
Affiliation(s)
- Melissa M Page
- Life Sciences Institute Diabetes Research Group and the Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada; Louvain Institute of Biomolecular Science and Technology, Université Catholique de Louvain, Louvain-la-Neuve, Belgium
| | - James D Johnson
- Life Sciences Institute Diabetes Research Group and the Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada. https://twitter.com/JimJohnsonSci
| |
Collapse
|
27
|
Arthur AE, Goss AM, Demark-Wahnefried W, Mondul AM, Fontaine KR, Chen YT, Carroll WR, Spencer SA, Rogers LQ, Rozek LS, Wolf GT, Gower BA. Higher carbohydrate intake is associated with increased risk of all-cause and disease-specific mortality in head and neck cancer patients: results from a prospective cohort study. Int J Cancer 2018; 143:1105-1113. [PMID: 29604042 DOI: 10.1002/ijc.31413] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 03/07/2018] [Accepted: 03/13/2018] [Indexed: 01/01/2023]
Abstract
No studies have evaluated associations between carbohydrate intake and head and neck squamous cell carcinoma (HNSCC) prognosis. We prospectively examined associations between pre- and post-treatment carbohydrate intake and recurrence, all-cause mortality, and HNSCC-specific mortality in a cohort of 414 newly diagnosed HNSCC patients. All participants completed pre- and post-treatment Food Frequency Questionnaires (FFQs) and epidemiologic surveys. Recurrence and mortality events were collected annually. Multivariable Cox Proportional Hazards models tested associations between carbohydrate intake (categorized into low, medium and high intake) and time to recurrence and mortality, adjusting for relevant covariates. During the study period, there were 70 deaths and 72 recurrences. In pretreatment analyses, high intakes of total carbohydrate (HR: 2.29; 95% CI: 1.23-4.25), total sugar (HR: 3.03; 95% CI: 1.12-3.68), glycemic load (HR: 2.10; 95% CI: 1.15-3.83) and simple carbohydrates (HR 2.26; 95% CI 1.19-4.32) were associated with significantly increased risk of all-cause mortality compared to low intake. High intakes of carbohydrate (HR 2.45; 95% CI: 1.23-4.25) and total sugar (HR 3.03; 95% CI 1.12-3.68) were associated with increased risk of HNSCC-specific mortality. In post-treatment analyses, medium fat intake was significantly associated with reduced risk of recurrence (HR 0.08; 95% CI 0.01-0.69) and all-cause mortality (HR 0.27; 95% CI 0.07-0.96). Stratification by tumor site and cancer stage in pretreatment analyses suggested effect modification by these factors. Our data suggest high pretreatment carbohydrate intake may be associated with adverse prognosis in HNSCC patients. Clinical intervention trials to further examine this hypothesis are warranted.
Collapse
Affiliation(s)
- Anna E Arthur
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL.,Carle Foundation Hospital, Carle Cancer Center, Urbana, IL
| | - Amy M Goss
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL
| | | | - Alison M Mondul
- Department of Epidemiology, University of Michigan, Ann Arbor, MI
| | - Kevin R Fontaine
- Department of Health Behavior, University of Alabama at Birmingham, Birmingham, AL
| | - Yi Tang Chen
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL
| | - William R Carroll
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, AL
| | - Sharon A Spencer
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, AL
| | - Laura Q Rogers
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL
| | - Laura S Rozek
- Department of Environmental Health Sciences, University of Michigan, Ann Arbor, MI.,Department of Otolaryngology, University of Michigan, Ann Arbor, MI
| | - Gregory T Wolf
- Department of Otolaryngology, University of Michigan, Ann Arbor, MI
| | - Barbara A Gower
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL
| | | |
Collapse
|
28
|
Gardner CD, Trepanowski JF, Del Gobbo LC, Hauser ME, Rigdon J, Ioannidis JPA, Desai M, King AC. Effect of Low-Fat vs Low-Carbohydrate Diet on 12-Month Weight Loss in Overweight Adults and the Association With Genotype Pattern or Insulin Secretion: The DIETFITS Randomized Clinical Trial. JAMA 2018; 319:667-679. [PMID: 29466592 PMCID: PMC5839290 DOI: 10.1001/jama.2018.0245] [Citation(s) in RCA: 394] [Impact Index Per Article: 65.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
IMPORTANCE Dietary modification remains key to successful weight loss. Yet, no one dietary strategy is consistently superior to others for the general population. Previous research suggests genotype or insulin-glucose dynamics may modify the effects of diets. OBJECTIVE To determine the effect of a healthy low-fat (HLF) diet vs a healthy low-carbohydrate (HLC) diet on weight change and if genotype pattern or insulin secretion are related to the dietary effects on weight loss. DESIGN, SETTING, AND PARTICIPANTS The Diet Intervention Examining The Factors Interacting with Treatment Success (DIETFITS) randomized clinical trial included 609 adults aged 18 to 50 years without diabetes with a body mass index between 28 and 40. The trial enrollment was from January 29, 2013, through April 14, 2015; the date of final follow-up was May 16, 2016. Participants were randomized to the 12-month HLF or HLC diet. The study also tested whether 3 single-nucleotide polymorphism multilocus genotype responsiveness patterns or insulin secretion (INS-30; blood concentration of insulin 30 minutes after a glucose challenge) were associated with weight loss. INTERVENTIONS Health educators delivered the behavior modification intervention to HLF (n = 305) and HLC (n = 304) participants via 22 diet-specific small group sessions administered over 12 months. The sessions focused on ways to achieve the lowest fat or carbohydrate intake that could be maintained long-term and emphasized diet quality. MAIN OUTCOMES AND MEASURES Primary outcome was 12-month weight change and determination of whether there were significant interactions among diet type and genotype pattern, diet and insulin secretion, and diet and weight loss. RESULTS Among 609 participants randomized (mean age, 40 [SD, 7] years; 57% women; mean body mass index, 33 [SD, 3]; 244 [40%] had a low-fat genotype; 180 [30%] had a low-carbohydrate genotype; mean baseline INS-30, 93 μIU/mL), 481 (79%) completed the trial. In the HLF vs HLC diets, respectively, the mean 12-month macronutrient distributions were 48% vs 30% for carbohydrates, 29% vs 45% for fat, and 21% vs 23% for protein. Weight change at 12 months was -5.3 kg for the HLF diet vs -6.0 kg for the HLC diet (mean between-group difference, 0.7 kg [95% CI, -0.2 to 1.6 kg]). There was no significant diet-genotype pattern interaction (P = .20) or diet-insulin secretion (INS-30) interaction (P = .47) with 12-month weight loss. There were 18 adverse events or serious adverse events that were evenly distributed across the 2 diet groups. CONCLUSIONS AND RELEVANCE In this 12-month weight loss diet study, there was no significant difference in weight change between a healthy low-fat diet vs a healthy low-carbohydrate diet, and neither genotype pattern nor baseline insulin secretion was associated with the dietary effects on weight loss. In the context of these 2 common weight loss diet approaches, neither of the 2 hypothesized predisposing factors was helpful in identifying which diet was better for whom. TRIAL REGISTRATION clinicaltrials.gov Identifier: NCT01826591.
Collapse
Affiliation(s)
- Christopher D. Gardner
- Stanford Prevention Research Center, Department of Medicine, Stanford University Medical School, Stanford, California
| | - John F. Trepanowski
- Stanford Prevention Research Center, Department of Medicine, Stanford University Medical School, Stanford, California
| | - Liana C. Del Gobbo
- Stanford Prevention Research Center, Department of Medicine, Stanford University Medical School, Stanford, California
| | - Michelle E. Hauser
- Stanford Prevention Research Center, Department of Medicine, Stanford University Medical School, Stanford, California
| | - Joseph Rigdon
- Quantitative Sciences Unit, Stanford University School of Medicine, Stanford, California
| | - John P. A. Ioannidis
- Stanford Prevention Research Center, Department of Medicine, Stanford University Medical School, Stanford, California
- Department of Health Research and Policy, Stanford University School of Medicine, Stanford, California
- Department of Statistics, Stanford University School of Humanities and Sciences, Stanford, California
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, California
| | - Manisha Desai
- Quantitative Sciences Unit, Stanford University School of Medicine, Stanford, California
- Department of Health Research and Policy, Stanford University School of Medicine, Stanford, California
- Department of Statistics, Stanford University School of Humanities and Sciences, Stanford, California
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, California
| | - Abby C. King
- Stanford Prevention Research Center, Department of Medicine, Stanford University Medical School, Stanford, California
- Department of Health Research and Policy, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
29
|
Ebbeling CB, Klein GL, Luoto PK, Wong JMW, Bielak L, Eddy RG, Steltz SK, Devlin C, Sandman M, Hron B, Shimy K, Heymsfield SB, Wolfe RR, Wong WW, Feldman HA, Ludwig DS. A randomized study of dietary composition during weight-loss maintenance: Rationale, study design, intervention, and assessment. Contemp Clin Trials 2018; 65:76-86. [PMID: 29233719 PMCID: PMC6055230 DOI: 10.1016/j.cct.2017.12.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 12/07/2017] [Accepted: 12/09/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND While many people with overweight or obesity can lose weight temporarily, most have difficulty maintaining weight loss over the long term. Studies of dietary composition typically focus on weight loss, rather than weight-loss maintenance, and rely on nutrition education and dietary counseling, rather than controlled feeding protocols. Variation in initial weight loss and insufficient differentiation among treatments confound interpretation of results and compromise conclusions regarding the weight-independent effects of dietary composition. The aim of the present study was to evaluate three test diets differing in carbohydrate-to-fat ratio during weight-loss maintenance. DESIGN AND DIETARY INTERVENTIONS Following weight loss corresponding to 12±2% of baseline body weight on a standard run-in diet, 164 participants aged 18 to 65years were randomly assigned to one of three test diets for weight-loss maintenance through 20weeks (test phase). We fed them high-carbohydrate (60% of energy from carbohydrate, 20% fat), moderate-carbohydrate (40% carbohydrate, 40% fat), and low-carbohydrate (20% carbohydrate, 60% fat) diets, controlled for protein content (20% of energy). During a 2-week ad libitum feeding phase following the test phase, we assessed the effect of the test diets on body weight. OUTCOMES The primary outcome was total energy expenditure, assessed by doubly-labeled water methodology. Secondary outcomes included resting energy expenditure and physical activity, chronic disease risk factors, and variables to inform an understanding of physiological mechanisms by which dietary carbohydrate-to-fat ratio might influence metabolism. Weight change during the ad libitum feeding phase was conceptualized as a proxy measure of hunger.
Collapse
Affiliation(s)
- Cara B Ebbeling
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States.
| | - Gloria L Klein
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Patricia K Luoto
- Department of Food and Nutrition, Framingham State University, 100 State Street, PO Box 9101, Framingham, MA 01701, United States
| | - Julia M W Wong
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Lisa Bielak
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Ralph G Eddy
- Sodexo Inc., Framingham State University, 100 State Street, PO Box 9101, Framingham, MA 01701, United States
| | - Sarah K Steltz
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Courtenay Devlin
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Megan Sandman
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Bridget Hron
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States; Division of Gastroenterology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Kim Shimy
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - Steven B Heymsfield
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States
| | - Robert R Wolfe
- University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - William W Wong
- Baylor College of Medicine, USDA/ARS Children's Nutrition Research Center, 1100 Bates Street, Houston, TX 77030, United States
| | - Henry A Feldman
- Institutional Centers for Clinical and Translational Research, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| | - David S Ludwig
- New Balance Foundation Obesity Prevention Center, Division of Endocrinology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, United States
| |
Collapse
|
30
|
Hron BM, Ebbeling CB, Feldman HA, Ludwig DS. Hepatic, adipocyte, enteric and pancreatic hormones: response to dietary macronutrient composition and relationship with metabolism. Nutr Metab (Lond) 2017; 14:44. [PMID: 28694840 PMCID: PMC5499060 DOI: 10.1186/s12986-017-0198-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 06/25/2017] [Indexed: 02/08/2023] Open
Abstract
Background We sought to characterize the effects of dietary macronutrient composition on various hormones implicated in the regulation of insulin sensitivity (IS) and energy expenditure (EE). Methods Following 10–15% weight loss, 21 overweight subjects consumed 3 weight-loss maintenance diets [low fat (LF), low glycemic index (LGI) and very low carbohydrate (VLC)] in random order, each for 4 weeks. At baseline and at the end of each treatment period, fasting samples for fibroblast growth factor (FGF)-21, heme-oxygenase-1 (HO-1), chemerin, irisin, secreted frizzle-related protein (SFRP-4), total bile acids, ghrelin, gastrin inhibitory peptide (GIP), peptide-Y, and amylin; hepatic and peripheral IS; and EE were obtained. Analyses were controlled for age, gender, baseline body mass index, and diet sequence. Results FGF-21 decreased (P < 0.0001), with differential effect by macronutrient composition (mean change from baseline ± SEM: LF −49.4 ± 16.6, LGI -58.6 ± 16.3, VLC -76.7 ± 18.2 pg/mL, P = 0.0002). Change in FGF-21 was inversely associated with change in hepatic IS [Beta = −0.565 units/log(ng/mL), P = 0.02], but not with peripheral IS or EE. Heme-oxygenase-1 (HO-1) increased (P = 0.003), without differential effect by macronutrient composition (LF 0.40 ± 0.26, LGI 0.98 ± 0.63, VLC 0.49 ± 0.29 ng/mL, P = 0.07). Ghrelin increased (P = 0.0003), while chemerin decreased (P = 0.001) without macronutrient effect. Total bile acid, irisin, SFRP-4, GIP, peptide-Y and amylin levels did not change. Conclusions FGF-21 levels decreased with dietary intervention in proportion to carbohydrate content, and correlated with hepatic insulin sensitivity, suggesting a pattern of improving FGF-21 resistance. HO-1 increased in response to dietary intervention, a tendency to greater increase in response to the LGI diet. Dietary intervention affected ghrelin and chemerin, independent of macronutrient composition. These findings may elucidate relationships between dietary composition, insulin sensitivity and metabolism. Trial registration NCT00315354.
Collapse
Affiliation(s)
- Bridget M Hron
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, 300 Longwood Ave., HUN Ground, Boston, MA USA.,New Balance Foundation Obesity Prevention Center and Division of Endocrinology, Boston Children's Hospital, 300 Longwood Ave, Boston, MA USA
| | - Cara B Ebbeling
- New Balance Foundation Obesity Prevention Center and Division of Endocrinology, Boston Children's Hospital, 300 Longwood Ave, Boston, MA USA
| | - Henry A Feldman
- Clinical Research Center, Boston Children's Hospital, 300 Longwood Ave, Boston, MA USA
| | - David S Ludwig
- New Balance Foundation Obesity Prevention Center and Division of Endocrinology, Boston Children's Hospital, 300 Longwood Ave, Boston, MA USA
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW This perspective is motivated by the need to question dogma that does not work: that the problem is insulin resistance (IR). We highlight the need to investigate potential environmental obesogens and toxins. RECENT FINDINGS The prequel to severe metabolic disease includes three interacting components that are abnormal: (a) IR, (b) elevated lipids and (c) elevated basal insulin (HI). HI is more common than IR and is a significant independent predictor of diabetes. We hypothesize that (1) the initiating defect is HI that increases nutrient consumption and hyperlipidemia (HL); (2) the cause of HI may include food additives, environmental obesogens or toxins that have entered our food supply since 1980; and (3) HI is sustained by HL derived from increased adipose mass and leads to IR. We suggest that HI and HL are early indicators of metabolic dysfunction and treating and reversing these abnormalities may prevent the development of more serious metabolic disease.
Collapse
Affiliation(s)
- Karel A. Erion
- 0000 0004 0367 5222grid.475010.7Obesity Research Center, Department of Medicine, Boston University School of Medicine, 650 Albany St, Boston, MA 02118 USA
- 0000 0000 9632 6718grid.19006.3eDivision of Endocrinology, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA USA
| | - Barbara E. Corkey
- 0000 0004 0367 5222grid.475010.7Obesity Research Center, Department of Medicine, Boston University School of Medicine, 650 Albany St, Boston, MA 02118 USA
| |
Collapse
|
32
|
van Rossum EFC. Obesity and cortisol: New perspectives on an old theme. Obesity (Silver Spring) 2017; 25:500-501. [PMID: 28229549 DOI: 10.1002/oby.21774] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Accepted: 01/03/2017] [Indexed: 12/31/2022]
Affiliation(s)
- Elisabeth F C van Rossum
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Obesity Center CGG, Rotterdam, the Netherlands
| |
Collapse
|
33
|
Camacho S, Ruppel A. Is the calorie concept a real solution to the obesity epidemic? Glob Health Action 2017; 10:1289650. [PMID: 28485680 PMCID: PMC5496172 DOI: 10.1080/16549716.2017.1289650] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 01/09/2017] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The obesity epidemic has been growing steadily across the whole world, and so far not a single country has been able to reverse it. The cause of obesity is stated by the World Health Organization as an energy imbalance between calories consumed and calories expended. However, growing evidence suggests that the calorie imbalance concept may not be sufficient to manage and reverse the obesity epidemic. OBJECTIVE To discuss the use of the calorie imbalance concept and its elements as a tool for weight management as well as its possible negative consequences and implications for public health, with the aim to point toward the need of an updated concept for causes of obesity. This update should guide public health interventions more efficiently to limit obesity by preventing weight gain or promoting weight loss. METHODS This is a literature reviews based on a semi-structured approach to determine the material to be examined. RESULTS After revisiting general facts about fat generation and accumulation, we propose an updated concept for the causes of obesity including diet composition and hormonal regulation of fat metabolism. CONCLUSIONS We discuss how this updated concept could benefit the overall efficiency of strategies against obesity, and hypothesize how potential resistance to adopting this new view could be lowered.
Collapse
Affiliation(s)
- Salvador Camacho
- Institute of Public Health, Universitätsklinikum Heidelberg, Heidelberg, Germany
- Alfred-Weber Institute, Universität Heidelberg, Heidelberg, Germany
| | - Andreas Ruppel
- Institute of Public Health, Universitätsklinikum Heidelberg, Heidelberg, Germany
| |
Collapse
|
34
|
Hall KD, Chen KY, Guo J, Leibel RL, Mayer LE, Reitman ML, Rosenbaum M, Smith SR, Walsh BT, Ravussin E. Reply to DS Ludwig and CB Ebbeling. Am J Clin Nutr 2016; 104:1488-1490. [PMID: 27802997 PMCID: PMC5081725 DOI: 10.3945/ajcn.116.143628] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Kevin D Hall
- From the National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD (KDH, e-mail: ; KYC; JG; MLR); Columbia University, New York, NY (RLL, LESM, MR, BTW); Pennington Biomedical Research Center, Baton Rouge, LA (ER); and The Translational Research Institute for Metabolism and Diabetes, Orlando, FL (SRS)
| | - Kong Y Chen
- From the National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD (KDH, e-mail: ; KYC; JG; MLR); Columbia University, New York, NY (RLL, LESM, MR, BTW); Pennington Biomedical Research Center, Baton Rouge, LA (ER); and The Translational Research Institute for Metabolism and Diabetes, Orlando, FL (SRS)
| | - Juen Guo
- From the National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD (KDH, e-mail: ; KYC; JG; MLR); Columbia University, New York, NY (RLL, LESM, MR, BTW); Pennington Biomedical Research Center, Baton Rouge, LA (ER); and The Translational Research Institute for Metabolism and Diabetes, Orlando, FL (SRS)
| | - Rudolph L Leibel
- From the National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD (KDH, e-mail: ; KYC; JG; MLR); Columbia University, New York, NY (RLL, LESM, MR, BTW); Pennington Biomedical Research Center, Baton Rouge, LA (ER); and The Translational Research Institute for Metabolism and Diabetes, Orlando, FL (SRS)
| | - Laurel Es Mayer
- From the National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD (KDH, e-mail: ; KYC; JG; MLR); Columbia University, New York, NY (RLL, LESM, MR, BTW); Pennington Biomedical Research Center, Baton Rouge, LA (ER); and The Translational Research Institute for Metabolism and Diabetes, Orlando, FL (SRS)
| | - Marc L Reitman
- From the National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD (KDH, e-mail: ; KYC; JG; MLR); Columbia University, New York, NY (RLL, LESM, MR, BTW); Pennington Biomedical Research Center, Baton Rouge, LA (ER); and The Translational Research Institute for Metabolism and Diabetes, Orlando, FL (SRS)
| | - Michael Rosenbaum
- From the National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD (KDH, e-mail: ; KYC; JG; MLR); Columbia University, New York, NY (RLL, LESM, MR, BTW); Pennington Biomedical Research Center, Baton Rouge, LA (ER); and The Translational Research Institute for Metabolism and Diabetes, Orlando, FL (SRS)
| | - Steven R Smith
- From the National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD (KDH, e-mail: ; KYC; JG; MLR); Columbia University, New York, NY (RLL, LESM, MR, BTW); Pennington Biomedical Research Center, Baton Rouge, LA (ER); and The Translational Research Institute for Metabolism and Diabetes, Orlando, FL (SRS)
| | - B Timothy Walsh
- From the National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD (KDH, e-mail: ; KYC; JG; MLR); Columbia University, New York, NY (RLL, LESM, MR, BTW); Pennington Biomedical Research Center, Baton Rouge, LA (ER); and The Translational Research Institute for Metabolism and Diabetes, Orlando, FL (SRS)
| | - Eric Ravussin
- From the National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD (KDH, e-mail: ; KYC; JG; MLR); Columbia University, New York, NY (RLL, LESM, MR, BTW); Pennington Biomedical Research Center, Baton Rouge, LA (ER); and The Translational Research Institute for Metabolism and Diabetes, Orlando, FL (SRS)
| |
Collapse
|
35
|
Affiliation(s)
- David S Ludwig
- From the New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Boston, MA (DSL, e-mail: ; CBE)
| | - Cara B Ebbeling
- From the New Balance Foundation Obesity Prevention Center, Boston Children's Hospital, Boston, MA (DSL, e-mail: ; CBE)
| |
Collapse
|
36
|
Ryan AS. Weight Regain Following Intentional Weight Loss in Older Adults. Curr Nutr Rep 2016. [DOI: 10.1007/s13668-016-0160-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|