251
|
Iwasaki S, Hamada T, Chisaki I, Andou T, Sano N, Furuta A, Amano N. Mechanism-Based Pharmacokinetic/Pharmacodynamic Modeling of the Glucagon-Like Peptide-1 Receptor Agonist Exenatide to Characterize Its Antiobesity Effects in Diet-Induced Obese Mice. J Pharmacol Exp Ther 2017; 362:441-449. [PMID: 28698254 DOI: 10.1124/jpet.117.242651] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 07/07/2017] [Indexed: 03/08/2025] Open
Abstract
In addition to their potent antidiabetic effects, glucagon-like peptide-1 (GLP-1) analogs lower body weight in humans. Hence, agonistic targeting of the GLP-1 receptor could be a valid approach to target obesity. However, quantitative analyses of the pharmacokinetic/pharmacodynamic (PK/PD) relationship between GLP-1 analogs and their antiobesity effect have not been reported in either animals or humans. Therefore, the present study was performed to establish a mechanism-based PK/PD model of GLP-1 receptor agonists using the GLP-1 analog exenatide for the development of promising new antiobesity drugs. Exenatide was administered to high-fat diet-induced obese C57BL/6J mice via subcutaneous bolus and continuous infusion. Food intake and body-weight reductions were observed and depended on the plasma concentrations of exenatide. The homeostatic feedback model, in which food intake is assumed to be regulated by appetite control signals, described the relationship among the plasma concentration-time profile of exenatide, food intake, and body weight. The estimated IC50 of exenatide against food intake was 2.05 pM, which is similar to the reported KD value of exenatide in rat brain and the estimated EC50 value for augmentation of insulin secretion in humans. The PK/PD model simulation indicated that subcutaneous infusion would show a stronger effect on body-weight reduction than bolus dosing would. This novel, quantitative PK/PD model could be used for antiobesity research and development of GLP-1 analogs, GLP-1 secretagogues, GLP-1 degradation inhibitors, and combinations thereof by allowing the estimation of appropriate pharmacokinetic profiles and dosing regimens.
Collapse
Affiliation(s)
- Shinji Iwasaki
- Drug Metabolism and Pharmacokinetics Research Laboratories (S.I., T.H., I.C., N.S., A.F., N.A.) and Integrated Technology Research Laboratories (T.A.), Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan
| | - Teruki Hamada
- Drug Metabolism and Pharmacokinetics Research Laboratories (S.I., T.H., I.C., N.S., A.F., N.A.) and Integrated Technology Research Laboratories (T.A.), Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan
| | - Ikumi Chisaki
- Drug Metabolism and Pharmacokinetics Research Laboratories (S.I., T.H., I.C., N.S., A.F., N.A.) and Integrated Technology Research Laboratories (T.A.), Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan
| | - Tomohiro Andou
- Drug Metabolism and Pharmacokinetics Research Laboratories (S.I., T.H., I.C., N.S., A.F., N.A.) and Integrated Technology Research Laboratories (T.A.), Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan
| | - Noriyasu Sano
- Drug Metabolism and Pharmacokinetics Research Laboratories (S.I., T.H., I.C., N.S., A.F., N.A.) and Integrated Technology Research Laboratories (T.A.), Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan
| | - Atsutoshi Furuta
- Drug Metabolism and Pharmacokinetics Research Laboratories (S.I., T.H., I.C., N.S., A.F., N.A.) and Integrated Technology Research Laboratories (T.A.), Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan
| | - Nobuyuki Amano
- Drug Metabolism and Pharmacokinetics Research Laboratories (S.I., T.H., I.C., N.S., A.F., N.A.) and Integrated Technology Research Laboratories (T.A.), Takeda Pharmaceutical Co., Ltd., Kanagawa, Japan
| |
Collapse
|
252
|
Buscail C, Sabate JM, Bouchoucha M, Torres MJ, Allès B, Hercberg S, Benamouzig R, Julia C. Association between self-reported vegetarian diet and the irritable bowel syndrome in the French NutriNet cohort. PLoS One 2017; 12:e0183039. [PMID: 28841661 PMCID: PMC5571937 DOI: 10.1371/journal.pone.0183039] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 07/29/2017] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND There is growing interest in using diet counselling in the management of patients with irritable bowel syndrome (IBS). Among new emerging diets, vegetarian diets (VD) seem to be experiencing an important popularity, partly because of their alleged health benefits. A recent study performed among a rural Indian population showed that predominant VD could be associated with IBS. OBJECTIVE This cross-sectional study aimed at assessing the association between the VD and IBS, among a large French cohort, the NutriNet-santé study. METHODS Subjects participating in the NutriNet-Santé cohort study completed a questionnaire based on Rome III criteria (N = 41,682). Anthropometrics, socio-demographical and lifestyle data, including VD, were collected prior to the completion of Rome III questionnaire via self-administered questionnaires. Association between VD and IBS and its subtypes was investigated through multivariate logistic regression. RESULTS The included subjects were mainly women (78.0%) and the mean age was 49.8±14.3 years. Among these individuals, 2,264 (5.4%) presented an IBS, and 805 (1.9%) reported a VD. Overall, VD was not associated with IBS or subtypes. A stable VD (i.e. self-declared at least three times) was associated with IBS (aOR 2.60 95%CI [1.37-4.91]), IBS mixed (aOR 2.97 95%CI [1.20-7.36]) and IBS diarrhoea (aOR 2.77 95%CI [1.01-7.59]). CONCLUSIONS This study suggests that a long term VD could be associated with IBS. Nevertheless, further studies are needed to confirm these results, and investigate the multiple aspects of the vegetarian diet, possibly related to the IBS.
Collapse
Affiliation(s)
- Camille Buscail
- Equipe de Recherche en Epidémiologie Nutritionnelle, Université Paris 13, Centre de Recherche en Epidémiologie et Biostatistiques (CRESS), Inserm 1153, Inra U1125, Cnam, COMUE Sorbonne Paris Cité, Bobigny, France
- Département de santé publique, Hôpital Avicenne, Bobigny, France
| | - Jean-Marc Sabate
- Service d’ Hépato-Gastro-Entérologie, Hôpital Avicenne, Bobigny, France
| | - Michel Bouchoucha
- Service d’ Hépato-Gastro-Entérologie, Hôpital Avicenne, Bobigny, France
| | - Marion J. Torres
- Equipe de Recherche en Epidémiologie Nutritionnelle, Université Paris 13, Centre de Recherche en Epidémiologie et Biostatistiques (CRESS), Inserm 1153, Inra U1125, Cnam, COMUE Sorbonne Paris Cité, Bobigny, France
| | - Benjamin Allès
- Equipe de Recherche en Epidémiologie Nutritionnelle, Université Paris 13, Centre de Recherche en Epidémiologie et Biostatistiques (CRESS), Inserm 1153, Inra U1125, Cnam, COMUE Sorbonne Paris Cité, Bobigny, France
| | - Serge Hercberg
- Equipe de Recherche en Epidémiologie Nutritionnelle, Université Paris 13, Centre de Recherche en Epidémiologie et Biostatistiques (CRESS), Inserm 1153, Inra U1125, Cnam, COMUE Sorbonne Paris Cité, Bobigny, France
- Département de santé publique, Hôpital Avicenne, Bobigny, France
| | - Robert Benamouzig
- Service d’ Hépato-Gastro-Entérologie, Hôpital Avicenne, Bobigny, France
| | - Chantal Julia
- Equipe de Recherche en Epidémiologie Nutritionnelle, Université Paris 13, Centre de Recherche en Epidémiologie et Biostatistiques (CRESS), Inserm 1153, Inra U1125, Cnam, COMUE Sorbonne Paris Cité, Bobigny, France
- Département de santé publique, Hôpital Avicenne, Bobigny, France
| |
Collapse
|
253
|
Bustos V, Partridge L. Good Ol' Fat: Links between Lipid Signaling and Longevity. Trends Biochem Sci 2017; 42:812-823. [PMID: 28802547 DOI: 10.1016/j.tibs.2017.07.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 07/07/2017] [Accepted: 07/13/2017] [Indexed: 12/13/2022]
Abstract
Aging is the single greatest risk factor for the development of disease. Understanding the biological molecules and mechanisms that modulate aging is therefore critical for the development of health-maximizing interventions for older people. The effect of fats on longevity has traditionally been disregarded as purely detrimental. However, new studies are starting to uncover the possible beneficial effects of lipids working as signaling molecules on health and longevity. These studies highlight the complex links between aging and lipid signaling. In this review we summarize accumulating evidence that points to changes in lipid metabolism, and in particular lipid signaling, as an underlying mechanism for healthy aging.
Collapse
Affiliation(s)
- Victor Bustos
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9b, 50931, Cologne, Germany
| | - Linda Partridge
- Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Strasse 9b, 50931, Cologne, Germany; Institute of Healthy Ageing, Department of Genetics, Evolution and Environment, University College London, Darwin Building, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
254
|
McDaid AF, Joshi PK, Porcu E, Komljenovic A, Li H, Sorrentino V, Litovchenko M, Bevers RPJ, Rüeger S, Reymond A, Bochud M, Deplancke B, Williams RW, Robinson-Rechavi M, Paccaud F, Rousson V, Auwerx J, Wilson JF, Kutalik Z. Bayesian association scan reveals loci associated with human lifespan and linked biomarkers. Nat Commun 2017; 8:15842. [PMID: 28748955 PMCID: PMC5537485 DOI: 10.1038/ncomms15842] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 05/08/2017] [Indexed: 02/07/2023] Open
Abstract
The enormous variation in human lifespan is in part due to a myriad of sequence variants, only a few of which have been revealed to date. Since many life-shortening events are related to diseases, we developed a Mendelian randomization-based method combining 58 disease-related GWA studies to derive longevity priors for all HapMap SNPs. A Bayesian association scan, informed by these priors, for parental age of death in the UK Biobank study (n=116,279) revealed 16 independent SNPs with significant Bayes factor at a 5% false discovery rate (FDR). Eleven of them replicate (5% FDR) in five independent longevity studies combined; all but three are depleted of the life-shortening alleles in older Biobank participants. Further analysis revealed that brain expression levels of nearby genes (RBM6, SULT1A1 and CHRNA5) might be causally implicated in longevity. Gene expression and caloric restriction experiments in model organisms confirm the conserved role for RBM6 and SULT1A1 in modulating lifespan.
Collapse
Affiliation(s)
- Aaron F McDaid
- Institute of Social and Preventive Medicine (IUMSP), Lausanne University Hospital, Lausanne 1010, Switzerland.,Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| | - Peter K Joshi
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AG, Scotland
| | - Eleonora Porcu
- Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland.,Center for Integrative Genomics, University of Lausanne, Lausanne 1015, Switzerland
| | - Andrea Komljenovic
- Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland.,Department of Ecology and Evolution, University of Lausanne, Lausanne 1015, Switzerland
| | - Hao Li
- Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Vincenzo Sorrentino
- Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Maria Litovchenko
- Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland.,Laboratory of Systems Biology and Genetics, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne and Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| | - Roel P J Bevers
- Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland.,Laboratory of Systems Biology and Genetics, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne and Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| | - Sina Rüeger
- Institute of Social and Preventive Medicine (IUMSP), Lausanne University Hospital, Lausanne 1010, Switzerland.,Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| | - Alexandre Reymond
- Center for Integrative Genomics, University of Lausanne, Lausanne 1015, Switzerland
| | - Murielle Bochud
- Institute of Social and Preventive Medicine (IUMSP), Lausanne University Hospital, Lausanne 1010, Switzerland
| | - Bart Deplancke
- Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland.,Laboratory of Systems Biology and Genetics, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne and Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| | - Robert W Williams
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | - Marc Robinson-Rechavi
- Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland.,Department of Ecology and Evolution, University of Lausanne, Lausanne 1015, Switzerland
| | - Fred Paccaud
- Institute of Social and Preventive Medicine (IUMSP), Lausanne University Hospital, Lausanne 1010, Switzerland
| | - Valentin Rousson
- Institute of Social and Preventive Medicine (IUMSP), Lausanne University Hospital, Lausanne 1010, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - James F Wilson
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Teviot Place, Edinburgh EH8 9AG, Scotland.,MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh EH4 2XU, Scotland
| | - Zoltán Kutalik
- Institute of Social and Preventive Medicine (IUMSP), Lausanne University Hospital, Lausanne 1010, Switzerland.,Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland
| |
Collapse
|
255
|
Saha SK, Lee SB, Won J, Choi HY, Kim K, Yang GM, Dayem AA, Cho SG. Correlation between Oxidative Stress, Nutrition, and Cancer Initiation. Int J Mol Sci 2017; 18:E1544. [PMID: 28714931 PMCID: PMC5536032 DOI: 10.3390/ijms18071544] [Citation(s) in RCA: 240] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 07/12/2017] [Accepted: 07/13/2017] [Indexed: 02/07/2023] Open
Abstract
Inadequate or excessive nutrient consumption leads to oxidative stress, which may disrupt oxidative homeostasis, activate a cascade of molecular pathways, and alter the metabolic status of various tissues. Several foods and consumption patterns have been associated with various cancers and approximately 30-35% of the cancer cases are correlated with overnutrition or malnutrition. However, several contradictory studies are available regarding the association between diet and cancer risk, which remains to be elucidated. Concurrently, oxidative stress is a crucial factor for cancer progression and therapy. Nutritional oxidative stress may be induced by an imbalance between antioxidant defense and pro-oxidant load due to inadequate or excess nutrient supply. Oxidative stress is a physiological state where high levels of reactive oxygen species (ROS) and free radicals are generated. Several signaling pathways associated with carcinogenesis can additionally control ROS generation and regulate ROS downstream mechanisms, which could have potential implications in anticancer research. Cancer initiation may be modulated by the nutrition-mediated elevation in ROS levels, which can stimulate cancer initiation by triggering DNA mutations, damage, and pro-oncogenic signaling. Therefore, in this review, we have provided an overview of the relationship between nutrition, oxidative stress, and cancer initiation, and evaluated the impact of nutrient-mediated regulation of antioxidant capability against cancer therapy.
Collapse
Affiliation(s)
- Subbroto Kumar Saha
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, Seoul 05029, Korea.
| | - Soo Bin Lee
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, Seoul 05029, Korea.
| | - Jihye Won
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, Seoul 05029, Korea.
| | - Hye Yeon Choi
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, Seoul 05029, Korea.
| | - Kyeongseok Kim
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, Seoul 05029, Korea.
| | - Gwang-Mo Yang
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, Seoul 05029, Korea.
| | - Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, Seoul 05029, Korea.
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Incurable Disease Animal Model & Stem Cell Institute (IDASI), Konkuk University, Seoul 05029, Korea.
| |
Collapse
|
256
|
Park JH, Yoo Y, Park YJ. Epigenetics: Linking Nutrition to Molecular Mechanisms in Aging. Prev Nutr Food Sci 2017; 22:81-89. [PMID: 28702424 PMCID: PMC5503416 DOI: 10.3746/pnf.2017.22.2.81] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/07/2017] [Indexed: 11/06/2022] Open
Abstract
Healthy aging has become a major goal of public health. Many studies have provided evidence and theories to explain molecular mechanisms of the aging process. Recent studies suggest that epigenetic mechanisms are responsible for life span and the progression of aging. Epigenetics is a fascinating field of molecular biology, which studies heritable modifications of DNA and histones that regulate gene expression without altering the DNA sequence. DNA methylation is a major epigenetic mark that shows progressive changes during aging. Recent studies have investigated aging-related DNA methylation as a biomarker that predicts cellular age. Interestingly, growing evidence proposes that nutrients play a crucial role in the regulation of epigenetic modifiers. Because various nutrients and their metabolites function as substrates or cofactors for epigenetic modifiers, nutrition can modulate or reverse epigenetic marks in the genome as well as expression patterns. Here, we will review the results on aging-associated epigenetic modifications and the possible mechanisms by which nutrition, including nutrient availability and bioactive compounds, regulate epigenetic changes and affect aging physiology.
Collapse
Affiliation(s)
- Joo Hyun Park
- Metabolism and Epigenetics Laboratory, Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea
| | - Yeongran Yoo
- Metabolism and Epigenetics Laboratory, Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea
| | - Yoon Jung Park
- Metabolism and Epigenetics Laboratory, Department of Nutritional Science and Food Management, Ewha Womans University, Seoul 03760, Korea
| |
Collapse
|
257
|
Thaiss CA, Shapiro H, Elinav E. Post-dieting weight gain: the role of persistent microbiome changes. Future Microbiol 2017; 12:555-559. [DOI: 10.2217/fmb-2017-0045] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Christoph A Thaiss
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Hagit Shapiro
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
258
|
Messaoudi I, Handu M, Rais M, Sureshchandra S, Park BS, Fei SS, Wright H, White AE, Jain R, Cameron JL, Winters-Stone KM, Varlamov O. Long-lasting effect of obesity on skeletal muscle transcriptome. BMC Genomics 2017; 18:411. [PMID: 28545403 PMCID: PMC5445270 DOI: 10.1186/s12864-017-3799-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 05/16/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Reduced physical activity and increased intake of calorically-dense diets are the main risk factors for obesity, glucose intolerance, and type 2 diabetes. Chronic overnutrition and hyperglycemia can alter gene expression, contributing to long-term obesity complications. While caloric restriction can reduce obesity and glucose intolerance, it is currently unknown whether it can effectively reprogram transcriptome to a pre-obesity level. The present study addressed this question by the preliminary examination of the transcriptional dynamics in skeletal muscle after exposure to overnutrition and following caloric restriction. RESULTS Six male rhesus macaques of 12-13 years of age consumed a high-fat western-style diet for 6 months and then were calorically restricted for 4 months without exercise. Skeletal muscle biopsies were subjected to longitudinal gene expression analysis using next-generation whole-genome RNA sequencing. In spite of significant weight loss and normalized insulin sensitivity, the majority of WSD-induced (n = 457) and WSD-suppressed (n = 47) genes remained significantly dysregulated after caloric restriction (FDR ≤0.05). The MetacoreTM pathway analysis reveals that western-style diet induced the sustained activation of the transforming growth factor-β gene network, associated with extracellular matrix remodeling, and the downregulation of genes involved in muscle structure development and nutritional processes. CONCLUSIONS Western-style diet, in the absence of exercise, induced skeletal muscle transcriptional programing, which persisted even after insulin resistance and glucose intolerance were completely reversed with caloric restriction.
Collapse
Affiliation(s)
- Ilhem Messaoudi
- School of Biological Sciences, University of California, Irvine, Irvine, CA, 92697, USA
| | - Mithila Handu
- Division of Cardiometabolic Health, Oregon National Primate Research Center, L584 505 NW 185th Ave., Beaverton, OR, 97006, USA
| | - Maham Rais
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, 92521, USA
| | - Suhas Sureshchandra
- School of Biological Sciences, University of California, Irvine, Irvine, CA, 92697, USA
| | - Byung S Park
- Department of Public Health and Preventive Medicine, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Suzanne S Fei
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
| | - Hollis Wright
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, 97006, USA
| | - Ashley E White
- Division of Cardiometabolic Health, Oregon National Primate Research Center, L584 505 NW 185th Ave., Beaverton, OR, 97006, USA
| | - Ruhee Jain
- Department of Neuroscience and Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Judy L Cameron
- Department of Neuroscience and Psychiatry, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Kerri M Winters-Stone
- Department of School of Nursing, Oregon Health and Science University, Portland, OR, 97239, USA
| | - Oleg Varlamov
- Division of Cardiometabolic Health, Oregon National Primate Research Center, L584 505 NW 185th Ave., Beaverton, OR, 97006, USA.
| |
Collapse
|
259
|
Micó V, Berninches L, Tapia J, Daimiel L. NutrimiRAging: Micromanaging Nutrient Sensing Pathways through Nutrition to Promote Healthy Aging. Int J Mol Sci 2017; 18:E915. [PMID: 28445443 PMCID: PMC5454828 DOI: 10.3390/ijms18050915] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Revised: 04/10/2017] [Accepted: 04/24/2017] [Indexed: 01/09/2023] Open
Abstract
Current sociodemographic predictions point to a demographic shift in developed and developing countries that will result in an unprecedented increase of the elderly population. This will be accompanied by an increase in age-related conditions that will strongly impair human health and quality of life. For this reason, aging is a major concern worldwide. Healthy aging depends on a combination of individual genetic factors and external environmental factors. Diet has been proved to be a powerful tool to modulate aging and caloric restriction has emerged as a valuable intervention in this regard. However, many questions about how a controlled caloric restriction intervention affects aging-related processes are still unanswered. Nutrient sensing pathways become deregulated with age and lose effectiveness with age. These pathways are a link between diet and aging. Thus, fully understanding this link is a mandatory step before bringing caloric restriction into practice. MicroRNAs have emerged as important regulators of cellular functions and can be modified by diet. Some microRNAs target genes encoding proteins and enzymes belonging to the nutrient sensing pathways and, therefore, may play key roles in the modulation of the aging process. In this review, we aimed to show the relationship between diet, nutrient sensing pathways and microRNAs in the context of aging.
Collapse
Affiliation(s)
- Víctor Micó
- Nutritional Genomics of Cardiovascular Disease and Obesity Fundation IMDEA Food, CEI UAM + CSIC, 28049 Madrid, Spain.
| | - Laura Berninches
- Nutritional Genomics of Cardiovascular Disease and Obesity Fundation IMDEA Food, CEI UAM + CSIC, 28049 Madrid, Spain.
| | - Javier Tapia
- Nutritional Genomics of Cardiovascular Disease and Obesity Fundation IMDEA Food, CEI UAM + CSIC, 28049 Madrid, Spain.
| | - Lidia Daimiel
- Nutritional Genomics of Cardiovascular Disease and Obesity Fundation IMDEA Food, CEI UAM + CSIC, 28049 Madrid, Spain.
- Department of Nutrition and Bromatology, CEU San Pablo University, Boadilla del Monte, 28668 Madrid, Spain.
| |
Collapse
|
260
|
Das SK, Roberts SB, Bhapkar MV, Villareal DT, Fontana L, Martin CK, Racette SB, Fuss PJ, Kraus WE, Wong WW, Saltzman E, Pieper CF, Fielding RA, Schwartz AV, Ravussin E, Redman LM. Body-composition changes in the Comprehensive Assessment of Long-term Effects of Reducing Intake of Energy (CALERIE)-2 study: a 2-y randomized controlled trial of calorie restriction in nonobese humans. Am J Clin Nutr 2017; 105:913-927. [PMID: 28228420 PMCID: PMC5366044 DOI: 10.3945/ajcn.116.137232] [Citation(s) in RCA: 85] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 01/19/2017] [Indexed: 01/28/2023] Open
Abstract
Background: Calorie restriction (CR) retards aging and increases longevity in many animal models. However, it is unclear whether CR can be implemented in humans without adverse effects on body composition.Objective: We evaluated the effect of a 2-y CR regimen on body composition including the influence of sex and body mass index (BMI; in kg/m2) among participants enrolled in CALERIE-2 (Comprehensive Assessment of Long-term Effects of Reducing Intake of Energy), a multicenter, randomized controlled trial.Design: Participants were 218 nonobese (BMI: 21.9-28.0) adults aged 21-51 y who were randomly assigned to 25% CR (CR, n = 143) or ad libitum control (AL, n = 75) in a 2:1 ratio. Measures at baseline and 12 and 24 mo included body weight, waist circumference, fat mass (FM), fat-free mass (FFM), and appendicular mass by dual-energy X-ray absorptiometry; activity-related energy expenditure (AREE) by doubly labeled water; and dietary protein intake by self-report. Values are expressed as means ± SDs.Results: The CR group achieved 11.9% ± 0.7% CR over 2-y and had significant decreases in weight (-7.6 ± 0.3 compared with 0.4 ± 0.5 kg), waist circumference (-6.2 ± 0.4 compared with 0.9 ± 0.5 cm), FM (-5.4 ± 0.3 compared with 0.5 ± 0.4 kg), and FFM (-2.0 ± 0.2 compared with -0.0 ± 0.2 kg) at 24 mo relative to the AL group (all between-group P < 0.001). Moreover, FFM as a percentage of body weight at 24 mo was higher, and percentage of FM was lower in the CR group than in the AL. AREE, but not protein intake, predicted preservation of FFM during CR (P < 0.01). Men in the CR group lost significantly more trunk fat (P = 0.03) and FFM expressed as a percentage of weight loss (P < 0.001) than women in the CR group.Conclusions: Two years of CR had broadly favorable effects on both whole-body and regional adiposity that could facilitate health span in humans. The decrements in FFM were commensurate with the reduced body mass; although men in the CR group lost more FFM than the women did, the percentage of FFM in the men in the CR group was higher than at baseline. CALERIE was registered at clinicaltrials.gov as NCT00427193.
Collapse
Affiliation(s)
- Sai Krupa Das
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA;
| | - Susan B Roberts
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - Manjushri V Bhapkar
- Duke Clinical Research Institute and Duke University Medical Center, Durham, NC
| | | | - Luigi Fontana
- CEINGE Biotecnologie Avanzate, Napoli, Italy;,Pennington Biomedical Research Center, Baton Rouge, LA;,USDA/Agricultural Research Services, Children’s Nutrition Research Center at Baylor College of Medicine, Houston, TX; and
| | | | | | - Paul J Fuss
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - William E Kraus
- Duke Clinical Research Institute and Duke University Medical Center, Durham, NC
| | - William W Wong
- USDA/Agricultural Research Services, Children’s Nutrition Research Center at Baylor College of Medicine, Houston, TX; and
| | - Edward Saltzman
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | - Carl F Pieper
- Duke Clinical Research Institute and Duke University Medical Center, Durham, NC
| | - Roger A Fielding
- Jean Mayer USDA Human Nutrition Research Center on Aging at Tufts University, Boston, MA
| | | | - Eric Ravussin
- Pennington Biomedical Research Center, Baton Rouge, LA
| | | | | |
Collapse
|
261
|
Marlatt KL, Redman LM, Burton JH, Martin CK, Ravussin E. Persistence of weight loss and acquired behaviors 2 y after stopping a 2-y calorie restriction intervention. Am J Clin Nutr 2017; 105:928-935. [PMID: 28275127 PMCID: PMC5366052 DOI: 10.3945/ajcn.116.146837] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 02/09/2017] [Indexed: 12/17/2022] Open
Abstract
Background: Calorie restriction (CR) influences aging processes and extends average and maximal life spans. The CALERIE 2 (Comprehensive Assessment of Long-Term Effects of Reducing Intake of Energy Phase 2) study was the first randomized clinical trial to examine the metabolic and psychological effects of CR in nonobese humans.Objective: We conducted a 2-y follow-up study of adults who underwent 2 y of CR or ad libitum (control) consumption and determined whether weight loss and acquired behaviors persisted after the study ended when participants determined their own lifestyle behaviors.Design: In this prospective, longitudinal study, we assessed differences in weight, body composition, psychological function, and energy expenditure in 39 nonobese [body mass index (in kg/m2): 22-28] men and women (25% CR: n = 24; control: n = 15) 12 and 24 mo after they completed the CALERIE 2 study at Pennington Biomedical.Results: Of 39 participants who were in the follow-up study, 29 subjects (CR: n = 18; control: n = 11) completed all visits at follow-up months 12 and 24. After the CR intervention, a mean ± SEM weight loss of 9.0 ± 0.6 kg was observed in the CR group, in which only 54% of the weight was regained 2 y later. Despite such a regain, weight, the percentage of body fat, and fat mass remained significantly reduced from baseline throughout follow-up and remained significantly less than in the control group (P < 0.05). At follow-up, the CR group retained higher degrees of dietary restraint and avoidance of certain foods.Conclusion: After a 2-y intensive CR intervention, ∼50% of CR-induced weight loss was maintained 2 y later, which was probably the result of lasting effects on acquired behaviors and dietary restraint. This trial was registered at clinicaltrials.gov as NCT00943215.
Collapse
Affiliation(s)
| | | | - Jeff H Burton
- Pennington Biomedical Research Center, Baton Rouge, LA
| | | | - Eric Ravussin
- Pennington Biomedical Research Center, Baton Rouge, LA
| |
Collapse
|
262
|
Molecular connections of obesity and aging: a focus on adipose protein 53 and retinoblastoma protein. Biogerontology 2017; 18:321-332. [PMID: 28357524 DOI: 10.1007/s10522-017-9698-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 03/27/2017] [Indexed: 12/13/2022]
Abstract
Obesity is an induced health problem that human beings have been facing with non-optimal treatment so far. Humans are on average getting fatter with age, and obesity and aging interact each other to shorten lifetime and decrease life quality. Obesity also causes several aging related-disorders such as cancer, strokes, cardiovascular disease, high blood pressure and type 2 diabetes. So, the molecular connections between aging and obesity are promising targets for bio-medical researches and innovative therapies of many health problems. In this review, we discuss the findings of adipose p53 and Rb-two central molecular linkages between aging and obesity-on lipid metabolism and obesity.
Collapse
|
263
|
Peterson CM, Zhang B, Johannsen DL, Ravussin E. Eight weeks of overfeeding alters substrate partitioning without affecting metabolic flexibility in men. Int J Obes (Lond) 2017; 41:887-893. [PMID: 28262678 PMCID: PMC5461218 DOI: 10.1038/ijo.2017.58] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Revised: 01/24/2017] [Accepted: 02/18/2017] [Indexed: 01/07/2023]
Abstract
Background/Objective Impairments in metabolic flexibility and substrate handling are associated with metabolic syndrome. However, it is unknown whether metabolic inflexibility causes insulin resistance. We therefore measured metabolic flexibility and substrate handling before and after 8 weeks of overfeeding in initially healthy adults, as a model of the early stages of insulin resistance. Subjects/Methods Twenty-nine healthy men (27 ± 5 years old; BMI 25.5 ± 2.3 kg/m2) were overfed by 40% above baseline energy requirements for 8 weeks and gained 7.6 ± 2.1 kg of weight. Before and after overfeeding, energy expenditure, substrate oxidation, and metabolic flexibility were measured in 2 ways: a) during 1 day of eucaloric feeding in a whole-room indirect calorimeter, and b) during a two-step hyperinsulinemic-euglycemic clamp. Results Eight weeks of overfeeding decreased insulin sensitivity at low and high doses of insulin (p=0.001 and p=0.06, respectively). This was accompanied by decreases in the respiratory quotient (RQ) while sleeping (0.877 ± 0.020 to 0.864 ± 0.026; p=0.05) and at low insulin levels during the clamp (0.927 ± 0.047 to 0.907 ± 0.032; p=0.01). Overfeeding did not affect metabolic flexibility as measured during a clamp (p≥0.17), but it tended to increase 24-hour metabolic flexibility (awake – sleep RQ) as measured by chamber by 0.010 ± 0.028 (p=0.08). In terms of substrate oxidation, overfeeding increased protein oxidation (13 ± 23 g/day; p=0.003) and tended to increase fat oxidation (6 ± 16 g/day; p=0.07), but did not affect carbohydrate oxidation (p=0.64). Individuals with greater metabolic adaptation to overfeeding had higher carbohydrate oxidation rates (r=0.66, p=8×10−5) but not fat oxidation rates (p=0.09). Conclusions The early stages of insulin resistance are accompanied by modest declines in the RQs during sleep and during a clamp, with no changes in fasting RQ or signs of metabolic inflexibility. Our data therefore suggest that metabolic inflexibility does not cause insulin resistance.
Collapse
Affiliation(s)
- C M Peterson
- Human Translational Physiology, Pennington Biomedical Research Center, Baton Rouge, LA, USA.,Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - B Zhang
- Human Translational Physiology, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - D L Johannsen
- Human Translational Physiology, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| | - E Ravussin
- Human Translational Physiology, Pennington Biomedical Research Center, Baton Rouge, LA, USA
| |
Collapse
|
264
|
Jorgensen A, Kalliokoski O, Forsberg K, Breitenstein K, Weimann A, Henriksen T, Hau J, Wörtwein G, Poulsen HE, Jorgensen MB. A chronic increase of corticosterone age-dependently reduces systemic DNA damage from oxidation in rats. Free Radic Biol Med 2017; 104:64-74. [PMID: 28069523 DOI: 10.1016/j.freeradbiomed.2017.01.013] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Revised: 01/04/2017] [Accepted: 01/06/2017] [Indexed: 11/26/2022]
Abstract
Stress and depression are associated with an acceleration of brain and bodily aging; effects which have been attributed to chronic elevations of glucocorticoids. We tested the hypothesis that a three week administration of stress-associated levels of corticosterone (CORT, the principal rodent glucocorticoid) would increase systemic and CNS DNA and RNA damage from oxidation; a phenomenon known to be centrally involved in the aging process. We also hypothesized that older individuals would be more sensitive to this effect and that the chronic CORT administration would exacerbate age-related memory decline. Young and old male Sprague-Dawley rats were non-invasively administered CORT by voluntary ingestion of nut paste containing either CORT (25mg/kg) or vehicle for a total of 22 days. CORT increased the 24h urinary excretion of the hormone to the levels previously observed after experimental psychological stress and caused a downregulation of the glucocorticoid receptor in the CA1 area of the hippocampus. Contrary to our hypothesis, 24h excretion of 8-oxodG/8-oxoGuo (markers of DNA/RNA damage from oxidation) was reduced in CORT-treated young animals, whereas old animals showed no significant differences. In old animals, CORT caused a borderline significant reduction of RNA oxidation in CNS, which was paralleled by a normalization of performance in an object location memory test. To our knowledge, this is the first demonstration that chronic stress-associated levels of CORT can reduce nucleic acid damage from oxidation. These findings contradict the notion of elevated CORT as a mediator of the accelerated aging observed in stress and depression.
Collapse
Affiliation(s)
- Anders Jorgensen
- Psychiatric Center Copenhagen (Rigshospitalet), Mental Health Services of the Capital Region of Denmark, Denmark; Laboratory of Neuropsychiatry, Psychiatric Center Copenhagen and Institute of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Denmark.
| | - Otto Kalliokoski
- Department of Experimental Medicine, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - Kristin Forsberg
- Laboratory of Neuropsychiatry, Psychiatric Center Copenhagen and Institute of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - Katrine Breitenstein
- Laboratory of Neuropsychiatry, Psychiatric Center Copenhagen and Institute of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - Allan Weimann
- Laboratory of Clinical Pharmacology, Copenhagen University Hospital Rigshospitalet, Denmark; Department of Clinical Pharmacology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Denmark
| | - Trine Henriksen
- Laboratory of Clinical Pharmacology, Copenhagen University Hospital Rigshospitalet, Denmark; Department of Clinical Pharmacology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Denmark
| | - Jann Hau
- Department of Experimental Medicine, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - Gitta Wörtwein
- Laboratory of Neuropsychiatry, Psychiatric Center Copenhagen and Institute of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Denmark
| | - Henrik Enghusen Poulsen
- Laboratory of Clinical Pharmacology, Copenhagen University Hospital Rigshospitalet, Denmark; Department of Clinical Pharmacology, Copenhagen University Hospital Bispebjerg and Frederiksberg, Denmark
| | - Martin Balslev Jorgensen
- Psychiatric Center Copenhagen (Rigshospitalet), Mental Health Services of the Capital Region of Denmark, Denmark; Laboratory of Neuropsychiatry, Psychiatric Center Copenhagen and Institute of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Denmark
| |
Collapse
|
265
|
Wade AT, Davis CR, Dyer KA, Hodgson JM, Woodman RJ, Keage HAD, Murphy KJ. A Mediterranean Diet to Improve Cardiovascular and Cognitive Health: Protocol for a Randomised Controlled Intervention Study. Nutrients 2017; 9:E145. [PMID: 28212320 PMCID: PMC5331576 DOI: 10.3390/nu9020145] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/24/2017] [Accepted: 02/09/2017] [Indexed: 02/07/2023] Open
Abstract
The Mediterranean diet has demonstrated efficacy for improving cardiovascular and cognitive health. However, a traditional Mediterranean diet delivers fewer serves of dairy and less dietary calcium than is currently recommended in Australia, which may limit long-term sustainability. The present study aims to evaluate whether a Mediterranean diet with adequate dairy and calcium can improve cardiovascular and cognitive function in an at-risk population, and thereby reduce risk of cardiovascular disease (CVD) and cognitive decline. A randomised, controlled, parallel, crossover design trial will compare a Mediterranean diet supplemented with dairy foods against a low-fat control diet. Forty participants with systolic blood pressure above 120 mmHg and at least two other risk factors of CVD will undertake each dietary intervention for eight weeks, with an eight-week washout period between interventions. Systolic blood pressure will be the primary measure of interest. Secondary outcomes will include measures of cardiometabolic health, dietary compliance, cognitive function, assessed using the Cambridge Neuropsychological Test Automated Battery (CANTAB), psychological well-being and dementia risk. This research will provide empirical evidence as to whether the Mediterranean diet can be modified to provide recommended dairy and calcium intakes while continuing to deliver positive effects for cardiovascular and cognitive health. The findings will hold relevance for the field of preventative healthcare and may contribute to revisions of national dietary guidelines.
Collapse
Affiliation(s)
- Alexandra T Wade
- Alliance for Research in Exercise, Nutrition and Activity, School of Health Sciences, University of South Australia, GPO Box 2471, Adelaide SA 5001, Australia.
| | - Courtney R Davis
- Alliance for Research in Exercise, Nutrition and Activity, School of Health Sciences, University of South Australia, GPO Box 2471, Adelaide SA 5001, Australia.
| | - Kathryn A Dyer
- Alliance for Research in Exercise, Nutrition and Activity, School of Health Sciences, University of South Australia, GPO Box 2471, Adelaide SA 5001, Australia.
| | - Jonathan M Hodgson
- School of Medicine and Pharmacology, Faculty of Medicine, Dentistry and Health Sciences, University of Western Australia, 35 Stirling Highway, Crawley WA 6009, Australia.
- School of Medical and Health Sciences, Edith Cowan University, Joondalup WA 6027, Australia.
| | - Richard J Woodman
- Flinders Centre for Epidemiology and Biostatistics, Flinders University, GPO Box 2100, Adelaide SA 5001, Australia.
| | - Hannah A D Keage
- Cognitive Ageing and Impairment Neurosciences, School of Psychology, Social Work and Social Policy, University of South Australia, GPO Box 2471, Adelaide SA 5001, Australia.
| | - Karen J Murphy
- Alliance for Research in Exercise, Nutrition and Activity, School of Health Sciences, University of South Australia, GPO Box 2471, Adelaide SA 5001, Australia.
| |
Collapse
|
266
|
Diamanti-Kandarakis E, Papalou O, Kandaraki EA, Kassi G. MECHANISMS IN ENDOCRINOLOGY: Nutrition as a mediator of oxidative stress in metabolic and reproductive disorders in women. Eur J Endocrinol 2017; 176:R79-R99. [PMID: 27678478 DOI: 10.1530/eje-16-0616] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2016] [Revised: 09/20/2016] [Accepted: 09/27/2016] [Indexed: 12/12/2022]
Abstract
Nutrition can generate oxidative stress and trigger a cascade of molecular events that can disrupt oxidative and hormonal balance. Nutrient ingestion promotes a major inflammatory and oxidative response at the cellular level in the postprandial state, altering the metabolic state of tissues. A domino of unfavorable metabolic changes is orchestrated in the main metabolic organs, including adipose tissue, skeletal muscle, liver and pancreas, where subclinical inflammation, endothelial dysfunction, mitochondrial deregulation and impaired insulin response and secretion take place. Simultaneously, in reproductive tissues, nutrition-induced oxidative stress can potentially violate delicate oxidative balance that is mandatory to secure normal reproductive function. Taken all the above into account, nutrition and its accompanying postprandial oxidative stress, in the unique context of female hormonal background, can potentially compromise normal metabolic and reproductive functions in women and may act as an active mediator of various metabolic and reproductive disorders.
Collapse
Affiliation(s)
| | - Olga Papalou
- Department of Endocrinology and Diabetes Center of ExcellenceEUROCLINIC, Athens, Greece
| | - Eleni A Kandaraki
- Endocrine Unit3rd Department of Internal Medicine, University of Athens Medical School, Athens, Greece
| | - Georgia Kassi
- Endocrine Unit3rd Department of Internal Medicine, University of Athens Medical School, Athens, Greece
| |
Collapse
|
267
|
Jongbloed F, Saat TC, Verweij M, Payan-Gomez C, Hoeijmakers JHJ, van den Engel S, van Oostrom CT, Ambagtsheer G, Imholz S, Pennings JLA, van Steeg H, IJzermans JNM, Dollé MET, de Bruin RWF. A signature of renal stress resistance induced by short-term dietary restriction, fasting, and protein restriction. Sci Rep 2017; 7:40901. [PMID: 28102354 PMCID: PMC5244361 DOI: 10.1038/srep40901] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 12/14/2016] [Indexed: 11/09/2022] Open
Abstract
During kidney transplantation, ischemia-reperfusion injury (IRI) induces oxidative stress. Short-term preoperative 30% dietary restriction (DR) and 3-day fasting protect against renal IRI. We investigated the contribution of macronutrients to this protection on both phenotypical and transcriptional levels. Male C57BL/6 mice were fed control food ad libitum, underwent two weeks of 30%DR, 3-day fasting, or received a protein-, carbohydrate- or fat-free diet for various periods of time. After completion of each diet, renal gene expression was investigated using microarrays. After induction of renal IRI by clamping the renal pedicles, animals were monitored seven days postoperatively for signs of IRI. In addition to 3-day fasting and two weeks 30%DR, three days of a protein-free diet protected against renal IRI as well, whereas the other diets did not. Gene expression patterns significantly overlapped between all diets except the fat-free diet. Detailed meta-analysis showed involvement of nuclear receptor signaling via transcription factors, including FOXO3, HNF4A and HMGA1. In conclusion, three days of a protein-free diet is sufficient to induce protection against renal IRI similar to 3-day fasting and two weeks of 30%DR. The elucidated network of common protective pathways and transcription factors further improves our mechanistic insight into the increased stress resistance induced by short-term DR.
Collapse
Affiliation(s)
- F Jongbloed
- Department of Surgery, Laboratory for Experimental Transplantation and Intestinal Surgery (LETIS), Erasmus University Medical Center, Rotterdam, the Netherlands.,Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - T C Saat
- Department of Surgery, Laboratory for Experimental Transplantation and Intestinal Surgery (LETIS), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - M Verweij
- Department of Surgery, Laboratory for Experimental Transplantation and Intestinal Surgery (LETIS), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - C Payan-Gomez
- Department of Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands.,Facultad de Ciencias Naturales y Matemáticas, Universidad del Rosario, Bogotá, Colombia
| | - J H J Hoeijmakers
- Department of Genetics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - S van den Engel
- Department of Surgery, Laboratory for Experimental Transplantation and Intestinal Surgery (LETIS), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - C T van Oostrom
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - G Ambagtsheer
- Department of Surgery, Laboratory for Experimental Transplantation and Intestinal Surgery (LETIS), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - S Imholz
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - J L A Pennings
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - H van Steeg
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands.,Department of Toxicogenetics, Leiden University Medical Center, Leiden, the Netherlands
| | - J N M IJzermans
- Department of Surgery, Laboratory for Experimental Transplantation and Intestinal Surgery (LETIS), Erasmus University Medical Center, Rotterdam, the Netherlands
| | - M E T Dollé
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - R W F de Bruin
- Department of Surgery, Laboratory for Experimental Transplantation and Intestinal Surgery (LETIS), Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
268
|
Mattison JA, Colman RJ, Beasley TM, Allison DB, Kemnitz JW, Roth GS, Ingram DK, Weindruch R, de Cabo R, Anderson RM. Caloric restriction improves health and survival of rhesus monkeys. Nat Commun 2017; 8:14063. [PMID: 28094793 PMCID: PMC5247583 DOI: 10.1038/ncomms14063] [Citation(s) in RCA: 585] [Impact Index Per Article: 73.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 11/24/2016] [Indexed: 12/23/2022] Open
Abstract
Caloric restriction (CR) without malnutrition extends lifespan and delays the onset of age-related disorders in most species but its impact in nonhuman primates has been controversial. In the late 1980s two parallel studies were initiated to determine the effect of CR in rhesus monkeys. The University of Wisconsin study reported a significant positive impact of CR on survival, but the National Institute on Aging study detected no significant survival effect. Here we present a direct comparison of longitudinal data from both studies including survival, bodyweight, food intake, fasting glucose levels and age-related morbidity. We describe differences in study design that could contribute to differences in outcomes, and we report species specificity in the impact of CR in terms of optimal onset and diet. Taken together these data confirm that health benefits of CR are conserved in monkeys and suggest that CR mechanisms are likely translatable to human health. Caloric restriction (CR) delays ageing of model organisms, but whether it works in nonhuman primates has been controversial. Here, the authors pool and reanalyse data from two long-running CR primate studies, concluding that moderate CR indeed improves health and survival of rhesus monkeys.
Collapse
Affiliation(s)
- Julie A Mattison
- Translational Gerontology Branch, National Institute on Aging, Baltimore, Maryland 21224, USA
| | - Ricki J Colman
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin 53715, USA
| | - T Mark Beasley
- Department of Biostatistics, University of Alabama, Birmingham, Alabama 35294, USA.,Geriatric Research Education and Clinical Center, Birmingham/Atlanta Veterans Administration Hospital, Birmingham, Alabama 35233, USA
| | - David B Allison
- Department of Biostatistics, University of Alabama, Birmingham, Alabama 35294, USA
| | - Joseph W Kemnitz
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, Wisconsin 53715, USA.,Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53792, USA
| | | | - Donald K Ingram
- Pennington Biomedical Research Center, Baton Rouge, Louisiana 70808, USA
| | - Richard Weindruch
- Department of Medicine, University of Wisconsin, Madison, Wisconsin 53792, USA.,Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, Baltimore, Maryland 21224, USA
| | - Rozalyn M Anderson
- Department of Medicine, University of Wisconsin, Madison, Wisconsin 53792, USA.,Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin 53705, USA
| |
Collapse
|
269
|
Sparks LM, Redman LM, Conley KE, Harper ME, Yi F, Hodges A, Eroshkin A, Costford SR, Gabriel ME, Shook C, Cornnell HH, Ravussin E, Smith SR. Effects of 12 Months of Caloric Restriction on Muscle Mitochondrial Function in Healthy Individuals. J Clin Endocrinol Metab 2017; 102:111-121. [PMID: 27778643 PMCID: PMC5413108 DOI: 10.1210/jc.2016-3211] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 10/21/2016] [Indexed: 11/19/2022]
Abstract
CONTEXT The effects of caloric restriction (CR) on in vivo muscle mitochondrial function in humans are controversial. OBJECTIVE We evaluated muscle mitochondrial function and associated transcriptional profiles in nonobese humans after 12 months of CR. DESIGN Individuals from an ancillary study of the CALERIE 2 randomized controlled trial were assessed at baseline and 12 months after a 25% CR or ad libitum (control) diet. SETTING The study was performed at Pennington Biomedical Research Center in Baton Rouge, LA. PARTICIPANTS Study participants included 51 (34 female subjects, 25 to 50 years of age) healthy nonobese individuals randomized to 1 of 2 groups (CR or control). INTERVENTION This study included 12 months of a 25% CR or ad libitum (control) diet. MAIN OUTCOMES In vivo mitochondrial function [maximal ATP synthesis rate (ATPmax), ATPflux/O2 (P/O)] was determined by 31P-magnetic resonance spectroscopy and optical spectroscopy, and body composition was determined by dual-energy X-ray absorptiometry. In a subset of individuals, a muscle biopsy was performed for transcriptional profiling via quantitative reverse transcription polymerase chain reaction and microarrays. RESULTS Weight, body mass index (BMI), fat, and fat-free mass (P < 0.001 for all) significantly decreased at month 12 after CR vs control. In vivo ATPmax and P/O were unaffected by 12 months of CR. Targeted transcriptional profiling showed no effects on pathways involved in mitochondrial biogenesis, function, or oxidative stress. A subgroup analysis according to baseline P/O demonstrated that a higher (vs lower) P/O was associated with notable improvements in ATPmax and P/O after CR. CONCLUSIONS In healthy nonobese humans, CR has no effect on muscle mitochondrial function; however, having a "more coupled" (versus "less coupled") phenotype enables CR-induced improvements in muscle mitochondrial function.
Collapse
Affiliation(s)
- Lauren M. Sparks
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida 32804;
- Clinical and Molecular Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827;
| | - Leanne M. Redman
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana 70808;
| | - Kevin E. Conley
- Radiology,
- Physiology & Biophysics, and
- Bioengineering, University of Washington Medical Center, Seattle, Washington 98195;
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada;
| | - Fanchao Yi
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida 32804;
| | - Andrew Hodges
- Bioinformatics Core, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and
| | - Alexey Eroshkin
- Bioinformatics Core, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and
| | | | - Meghan E. Gabriel
- Clinical and Molecular Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827;
| | - Cherie Shook
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida 32804;
| | - Heather H. Cornnell
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida 32804;
| | - Eric Ravussin
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana 70808;
| | - Steven R. Smith
- Translational Research Institute for Metabolism and Diabetes, Florida Hospital, Orlando, Florida 32804;
- Clinical and Molecular Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827;
| |
Collapse
|
270
|
Broderick TL, Jankowski M, Gutkowska J. The effects of exercise training and caloric restriction on the cardiac oxytocin natriuretic peptide system in the diabetic mouse. Diabetes Metab Syndr Obes 2017; 10:27-36. [PMID: 28138261 PMCID: PMC5238809 DOI: 10.2147/dmso.s115453] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Regular exercise training (ET) and caloric restriction (CR) are the frontline strategies in the treatment of type 2 diabetes mellitus with the aim at reducing cardiometabolic risk. ET and CR improve body weight and glycemic control, and experimental studies indicate that these paradigms afford cardioprotection. In this study, the effects of combined ET and CR on the cardioprotective oxytocin (OT)-natriuretic peptide (NP) system were determined in the db/db mouse, a model of type 2 diabetes associated with insulin resistance, hyperglycemia, and obesity. METHODS Five-week-old male db/db mice were assigned to the following groups: sedentary, ET, and ET + CR. Nonobese heterozygote littermates served as controls. ET was performed on a treadmill at moderate intensity, and CR was induced by reducing food intake by 30% of that consumed by sedentary db/db mice for a period of 8 weeks. RESULTS After 8 weeks, only ET + CR, but not ET, slightly improved body weight compared to sedentary db/db mice. Regardless of the treatment, db/db mice remained hyperglycemic. Hearts from db/db mice demonstrated reduced expression of genes linked to the cardiac OT-NP system. In fact, compared to control mice, mRNA expression of GATA binding protein 4 (GATA4), OT receptor, OT, brain NP, NP receptor type C, and endothelial nitric oxide synthase (eNOS) was decreased in hearts from sedentary db/db mice. Both ET alone and ET + CR increased the mRNA expression of GATA4 compared to sedentary db/db mice. Only ET combined with CR produced increased eNOS mRNA and protein expression. CONCLUSION Our data indicate that enhancement of eNOS by combined ET and CR may improve coronary endothelial vasodilator dysfunction in type 2 diabetes but did not prevent the downregulation of cardiac expression in the OT-NP system, possibly resulting from the sustained hyperglycemia and obesity in diabetic mice.
Collapse
Affiliation(s)
- Tom L Broderick
- Department of Physiology, Laboratory of Diabetes and Exercise Metabolism, Midwestern University, Glendale, AZ, USA
- Correspondence: Tom L Broderick, Department of Physiology, Laboratory of Diabetes and Exercise Metabolism, Midwestern University, 19555 North 59th Avenue, Glendale, AZ 85308, USA, Tel +1 623 572 3664, Fax +1 623 572 3673, Email
| | - Marek Jankowski
- Department of Medicine, Laboratory of Cardiovascular Biochemistry, Centre Hospitalier de l‘Université de Montréal-Hôtel-Dieu, Montréal, QC, Canada
| | - Jolanta Gutkowska
- Department of Medicine, Laboratory of Cardiovascular Biochemistry, Centre Hospitalier de l‘Université de Montréal-Hôtel-Dieu, Montréal, QC, Canada
| |
Collapse
|
271
|
Himbert C, Thompson H, Ulrich CM. Effects of Intentional Weight Loss on Markers of Oxidative Stress, DNA Repair and Telomere Length - a Systematic Review. Obes Facts 2017; 10:648-665. [PMID: 29237161 PMCID: PMC5836214 DOI: 10.1159/000479972] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 07/27/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Altered levels of markers of oxidative stress, DNA repair, and telomere integrity have been detected in obese individuals and may underlie the pathogenesis of obesity-related diseases. However, whether or not such effects are reversed by intentional weight loss has not been systematically reviewed. METHODS A literature search in PubMed/Medline identified 2,388 articles of which 21 studies (randomized controlled trial (RCT) (n = 10) and non-randomized intervention studies (n = 11)) were classified as testing the effects of intentional weight loss on i) oxidative stress (n = 15), ii) DNA repair (n = 2), and iii) telomere length (n = 4). RESULTS Across a broad range of intervention designs, diet-, exercise-, surgery-, balloon-induced weight loss regimens decreased oxidative stress measures. Studies investigating DNA repair capacity or telomere length as endpoints after weight loss were less common in number and yielded null or inconsistent results, respectively. CONCLUSION While this systematic review supports a role for intentional weight loss in reducing obesity-associated oxidative stress, it is not clear whether the effects are primary outcomes or secondary to improvement in obesity-associated insulin resistance and/or chronic inflammation. Although the lack of effect of intentional weight loss on DNA repair capacity might be anticipated given that oxidative stress is reduced, additional studies are needed. The inconsistent effects of weight loss on telomere length or DNA repair suggest the need for a re-assessment of intervention designs and assay methodology to definitively address this topic.
Collapse
Affiliation(s)
- Caroline Himbert
- Department of Population Sciences, Huntsman Cancer Institute, Salt Lake City, UT, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
- University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Henry Thompson
- Cancer Prevention Laboratory, Colorado State University, Fort Collins, CO, USA
| | - Cornelia M. Ulrich
- Department of Population Sciences, Huntsman Cancer Institute, Salt Lake City, UT, USA
- Department of Population Health Sciences, University of Utah, Salt Lake City, UT, USA
- *Cornelia M. Ulrich, Department of Population Sciences, Huntsman Cancer Institute, 2000 Circle of Hope Drive, Room 4725, Salt Lake City, UT 84112, USA,
| |
Collapse
|
272
|
Weiss EP, Jordan RC, Frese EM, Albert SG, Villareal DT. Effects of Weight Loss on Lean Mass, Strength, Bone, and Aerobic Capacity. Med Sci Sports Exerc 2017; 49:206-217. [PMID: 27580151 PMCID: PMC5161655 DOI: 10.1249/mss.0000000000001074] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE This study aimed to evaluate the hypothesis that exercise attenuates the reductions in lean mass, muscle strength, bone mineral density, and V˙O2max that accompany modest weight loss induced by calorie restriction (CR). METHODS Overweight, sedentary women and men (n = 52, 45-65 yr) were randomized to 6%-8% weight loss by using CR, endurance exercise training (EX), or both (CREX). The CR and the CREX groups underwent counseling to reduce energy intake by 20% and 10%, respectively. The EX and the CREX groups exercised 7.4 ± 0.5 and 4.4 ± 0.5 h·wk, respectively. Before and after 16.8 ± 1.1 wk of weight loss, lean mass and bone mineral density were measured with dual-energy x-ray absorptiometry, strength was measured with dynamometry, and aerobic capacity (V˙O2max) was measured with indirect calorimetry during maximal-intensity treadmill exercise. RESULTS Weight loss was ~7% in all groups. Decreases in whole-body (~2%, P = 0.003) and lower extremity (~4%, P < 0.0001) lean mass occurred in the CR group (both P < 0.05). Despite similar weight loss, these reductions were attenuated in the CREX group (~1%, P = 0.44 and ~2%, P = 0.05, respectively) and absent in the EX group. Absolute aerobic capacity decreased ~6% in the CR group (P = 0.04), was unchanged in the CREX group (P = 0.28), and increased ~15% in the EX group (P < 0.0001). No changes in muscle strength or bone were observed. CONCLUSIONS Modest weight loss (~7%) induced by 20% CR in overweight women and men decreases lean mass and reduces absolute V˙O2max. Exercise protects against these effects. Although the CR-induced changes might be considered physiologically appropriate for a reduced body weight, exercise preserves and/or improves these parameters during weight loss, which likely improves physical function. These findings support the notion of using exercise as an important component of weight loss programs.
Collapse
Affiliation(s)
- Edward P. Weiss
- Department of Nutrition and Dietetics, Saint Louis University, St. Louis, MO
- Division of Geriatrics and Nutritional Science, Washington University School of Medicine, St. Louis, MO
| | - Richard C. Jordan
- Department of Nutrition and Dietetics, Saint Louis University, St. Louis, MO
| | - Ethel M. Frese
- Department of Physical Therapy and Athletic Training, Saint Louis University, St. Louis, MO
| | - Stewart G. Albert
- Division of Endocrinology, School of Medicine, Saint Louis University, St. Louis, MO
| | - Dennis T. Villareal
- Division of Geriatrics and Nutritional Science, Washington University School of Medicine, St. Louis, MO
- Division of Endocrinology, Diabetes, and Metabolism, Baylor College of Medicine and Michael E DeBakey VA Medical Center, Houston, TX
| |
Collapse
|
273
|
Beck WR, Scariot PPM, Carmo SSD, Manchado-Gobatto FB, Gobatto CA. Metabolic profile and spontaneous physical activity modulation under short-term food restriction in young rats. MOTRIZ: REVISTA DE EDUCACAO FISICA 2017. [DOI: 10.1590/s1980-6574201700si0013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
274
|
Use of Salivary Diurnal Cortisol as an Outcome Measure in Randomised Controlled Trials: a Systematic Review. Ann Behav Med 2016; 50:210-36. [PMID: 27007274 PMCID: PMC4823366 DOI: 10.1007/s12160-015-9753-9] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background Dysregulation of the hypothalamic-pituitary-adrenal (HPA) axis is associated with diverse adverse health outcomes, making it an important therapeutic target. Measurement of the diurnal rhythm of cortisol secretion provides a window into this system. At present, no guidelines exist for the optimal use of this biomarker within randomised controlled trials (RCTs). Purpose The aim of this study is to describe the ways in which salivary diurnal cortisol has been measured within RCTs of health or behavioural interventions in adults. Methods Six electronic databases (up to May 21, 2015) were systematically searched for RCTs which used salivary diurnal cortisol as an outcome measure to evaluate health or behavioural interventions in adults. A narrative synthesis was undertaken of the findings in relation to salivary cortisol methodology and outcomes. Results From 78 studies that fulfilled the inclusion criteria, 30 included healthy participants (38.5 %), 27 included patients with physical disease (34.6 %) and 21 included patients with psychiatric disease (26.9 %). Psychological therapies were most commonly evaluated (n = 33, 42.3 %). There was substantial heterogeneity across studies in relation to saliva collection protocols and reported cortisol parameters. Only 39 studies (50 %) calculated a rhythm parameter such as the diurnal slope or the cortisol awakening response (CAR). Patterns of change in cortisol parameters were inconsistent both within and across studies and there was low agreement with clinical findings. Conclusions Salivary diurnal cortisol is measured inconsistently across RCTs, which is limiting the interpretation of findings within and across studies. This indicates a need for more validation work, along with consensus guidelines. Electronic supplementary material The online version of this article (doi:10.1007/s12160-015-9753-9) contains supplementary material, which is available to authorized users.
Collapse
|
275
|
Ten Haaf T, Verreijen AM, Memelink RG, Tieland M, Weijs PJM. Reduction in energy expenditure during weight loss is higher than predicted based on fat free mass and fat mass in older adults. Clin Nutr 2016; 37:250-253. [PMID: 28062083 DOI: 10.1016/j.clnu.2016.12.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 12/09/2016] [Accepted: 12/19/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND & AIM The aim of this study was to describe a decrease in resting energy expenditure during weight loss that is larger than expected based on changes in body composition, called adaptive thermogenesis (AT), in overweight and obese older adults. METHODS Multiple studies were combined to assess AT in younger and older subjects. Body composition and resting energy expenditure (REE) were measured before and after weight loss. Baseline values were used to predict fat free mass and fat mass adjusted REE after weight loss. AT was defined as the difference between predicted and measured REE after weight loss. The median age of 55 y was used as a cutoff to compare older with younger subjects. The relation between AT and age was investigated using linear regression analysis. RESULTS In this study 254 (M = 88, F = 166) overweight and obese subjects were included (BMI: 31.7 ± 4.4 kg/m2, age: 51 ± 14 y). The AT was only significant for older subjects (64 ± 185 kcal/d, 95% CI [32, 96]), but not for younger subjects (19 ± 152 kcal/d, 95% CI [-9, 46]). The size of the AT was significantly higher for older compared to younger adults (β = 47, p = 0.048), independent of gender and type and duration of the weight loss program. CONCLUSIONS We conclude that adaptive thermogenesis is present only in older subjects, which might have implications for weight management in older adults. A reduced energy intake is advised to counteract the adaptive thermogenesis.
Collapse
Affiliation(s)
- Twan Ten Haaf
- Department of Nutrition and Dietetics, School of Sports and Nutrition, Amsterdam University of Applied Sciences, Dr. Meurerlaan 8, 1067SM, Amsterdam, The Netherlands
| | - Amely M Verreijen
- Department of Nutrition and Dietetics, School of Sports and Nutrition, Amsterdam University of Applied Sciences, Dr. Meurerlaan 8, 1067SM, Amsterdam, The Netherlands
| | - Robert G Memelink
- Department of Nutrition and Dietetics, School of Sports and Nutrition, Amsterdam University of Applied Sciences, Dr. Meurerlaan 8, 1067SM, Amsterdam, The Netherlands
| | - Michael Tieland
- Department of Nutrition and Dietetics, School of Sports and Nutrition, Amsterdam University of Applied Sciences, Dr. Meurerlaan 8, 1067SM, Amsterdam, The Netherlands
| | - Peter J M Weijs
- Department of Nutrition and Dietetics, School of Sports and Nutrition, Amsterdam University of Applied Sciences, Dr. Meurerlaan 8, 1067SM, Amsterdam, The Netherlands; Department of Nutrition and Dietetics, Internal Medicine, VU University Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
276
|
van de Rest O, Schutte BAM, Deelen J, Stassen SAM, van den Akker EB, van Heemst D, Dibbets-Schneider P, van Dipten-van der Veen RA, Kelderman M, Hankemeier T, Mooijaart SP, van der Grond J, Houwing-Duistermaat JJ, Beekman M, Feskens EJM, Slagboom PE. Metabolic effects of a 13-weeks lifestyle intervention in older adults: The Growing Old Together Study. Aging (Albany NY) 2016; 8:111-26. [PMID: 26824634 PMCID: PMC4761717 DOI: 10.18632/aging.100877] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
For people in their 40s and 50s, lifestyle programs have been shown to improve metabolic health. For older adults, however, it is not clear whether these programs are equally healthy. In the Growing Old Together study, we applied a 13-weeks lifestyle program, with a target of 12.5% caloric restriction and 12.5% increase in energy expenditure through an increase in physical activity, in 164 older adults (mean age=63.2 years; BMI=23-35 kg/m2). Mean weight loss was 4.2% (SE=2.8%) of baseline weight, which is comparable to a previous study in younger adults. Fasting insulin levels, however, showed a much smaller decrease (0.30 mU/L (SE=3.21)) and a more heterogeneous response (range=2.0-29.6 mU/L). Many other parameters of metabolic health, such as blood pressure, and thyroid, glucose and lipid metabolism improved significantly. Many 1H-NMR metabolites changed in a direction previously associated with a low risk of type 2 diabetes and cardiovascular disease and partially independently of weight loss. In conclusion, 25% reduction in energy balance for 13 weeks induced a metabolic health benefit in older adults, monitored by traditional and novel metabolic markers.
Collapse
Affiliation(s)
- Ondine van de Rest
- Division of Human Nutrition, Wageningen University, 6700 EV Wageningen, The Netherlands
| | - Bianca A M Schutte
- Department of Molecular Epidemiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Joris Deelen
- Department of Molecular Epidemiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Stephanie A M Stassen
- Department of Gerontology and Geriatrics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Erik B van den Akker
- Department of Molecular Epidemiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands.,The Delft Bioinformatics Lab, Delft University of Technology, 2628 CD Delft, The Netherlands
| | - Diana van Heemst
- Department of Gerontology and Geriatrics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | | | | | - Milou Kelderman
- Division of Human Nutrition, Wageningen University, 6700 EV Wageningen, The Netherlands
| | - Thomas Hankemeier
- Division of Analytical Biosciences, Leiden Academic Centre for Drug Research, Leiden University, Leiden 2300 RA, The Netherlands
| | - Simon P Mooijaart
- Department of Gerontology and Geriatrics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Jeroen van der Grond
- Department of Radiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | | | - Marian Beekman
- Department of Molecular Epidemiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Edith J M Feskens
- Division of Human Nutrition, Wageningen University, 6700 EV Wageningen, The Netherlands
| | - P Eline Slagboom
- Department of Molecular Epidemiology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| |
Collapse
|
277
|
Sparks LM, Redman LM, Conley KE, Harper ME, Hodges A, Eroshkin A, Costford SR, Gabriel ME, Yi F, Shook C, Cornnell HH, Ravussin E, Smith SR. Differences in Mitochondrial Coupling Reveal a Novel Signature of Mitohormesis in Muscle of Healthy Individuals. J Clin Endocrinol Metab 2016; 101:4994-5003. [PMID: 27710240 PMCID: PMC5155692 DOI: 10.1210/jc.2016-2742] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
CONTEXT Reduced mitochondrial coupling (ATP/O2 [P/O]) is associated with sedentariness and insulin resistance. Interpreting the physiological relevance of P/O measured in vitro is challenging. OBJECTIVE To evaluate muscle mitochondrial function and associated transcriptional profiles in nonobese healthy individuals distinguished by their in vivo P/O. DESIGN Individuals from an ancillary study of Comprehensive Assessment of Long-term Effects of Reducing Intake of Energy phase 2 were assessed at baseline. SETTING The study was performed at Pennington Biomedical Research Center. PARTICIPANTS Forty-seven (18 males, 26-50 y of age) sedentary, healthy nonobese individuals were divided into 2 groups based on their in vivo P/O. INTERVENTION None. Main Outcome(s): Body composition by dual-energy x-ray absorptiometry, in vivo mitochondrial function (P/O and maximal ATP synthetic capacity) by 31P-magnetic resonance spectroscopy and optical spectroscopy were measured. A muscle biopsy was performed to measure fiber type, transcriptional profiling (microarray), and protein expressions. RESULTS No differences in body composition, peak aerobic capacity, type I fiber content, or mitochondrial DNA copy number were observed between the 2 groups. Compared with the uncoupled group (lower P/O), the coupled group (higher P/O) had higher rates of maximal ATP synthetic capacity (maximal ATP synthetic capacity, P < .01). Transcriptomics analyses revealed higher expressions of genes involved in mitochondrial remodeling and the oxidative stress response in the coupled group. A trend for higher mitonuclear protein imbalance (P = .06) and an elevated mitochondrial unfolded protein response (heat shock protein 60 protein; P = .004) were also identified in the coupled group. CONCLUSIONS Higher muscle mitochondrial coupling is accompanied by an overall elevation in mitochondrial function, a novel transcriptional signature of oxidative stress and mitochondrial remodeling and indications of an mitochondrial unfolded protein response.
Collapse
Affiliation(s)
- Lauren M Sparks
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Leanne M Redman
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Kevin E Conley
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Mary-Ellen Harper
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Andrew Hodges
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Alexey Eroshkin
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Sheila R Costford
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Meghan E Gabriel
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Fanchao Yi
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Cherie Shook
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Heather H Cornnell
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Eric Ravussin
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| | - Steven R Smith
- Translational Research Institute for Metabolism and Diabetes (L.M.S., F.Y., C.S., H.H.C., S.R.S.), Florida Hospital, Orlando, Florida 32804; Clinical and Molecular Origins of Disease (L.M.S., M.E.G., S.R.S.), Sanford Burnham Prebys Medical Discovery Institute, Orlando, Florida 32827; Pennington Biomedical Research Center (L.M.R., E.R.), Louisiana State University System, Baton Rouge, Louisiana 70808; Departments of Radiology, Physiology and Biophysics, and Bioengineering (K.E.C.), University of Washington Medical Center, Seattle, Washington 98195; Department of Biochemistry, Microbiology, and Immunology (M.-E.H.), University of Ottawa, Ottawa, Ontario ON K1N 6N5, Canada; Bioinformatics Core (A.H., A.E.), Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California 92037; and Hospital for Sick Children (S.R.C.), Toronto, Ontario, ON M5G 1X8 Canada
| |
Collapse
|
278
|
Core body temperature is lower in postmenopausal women than premenopausal women: potential implications for energy metabolism and midlife weight gain. Cardiovasc Endocrinol 2016; 5:151-154. [PMID: 28111609 DOI: 10.1097/xce.0000000000000078] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
OBJECTIVE Weight gain during the menopausal transition is common. Although studies have suggested that weight gain is more likely related to aging than menopause, there is a reduction in resting energy expenditure with surgical or natural menopause which is independent of age and changes in body composition. The underlying mechanisms could include a reduction in core body temperature. METHODS Data were obtained from two related studies. Sample size was 23 men and 25 women (12 premenopausal,13 postmenopausal). In the Clinical Research Unit, core temperature was measured every minute for 24 hours (CorTemp System,HQ Inc.). RESULTS Mean 24-hour core body temperature was 0.25 ± 0.06 °C lower in postmenopausal than premenopausal women (p=0.001). Mean 24-hour core temperature was 0.34 ± 0.05 °C lower in men than in premenopausal women (p<0.001). CONCLUSIONS Postmenopausal women, like men, had lower core body temperatures than premenopausal women. This may have implications for midlife weight gain.
Collapse
|
279
|
Simonsick EM, Meier HCS, Shaffer NC, Studenski SA, Ferrucci L. Basal body temperature as a biomarker of healthy aging. AGE (DORDRECHT, NETHERLANDS) 2016; 38:445-454. [PMID: 27785691 PMCID: PMC5266228 DOI: 10.1007/s11357-016-9952-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 10/14/2016] [Indexed: 05/29/2023]
Abstract
Scattered evidence indicates that a lower basal body temperature may be associated with prolonged health span, yet few studies have directly evaluated this relationship. We examined cross-sectional and longitudinal associations between early morning oral temperature (95.0-98.6 °F) and usual gait speed, endurance walk performance, fatigability, and grip strength in 762 non-frail men (52 %) and women aged 65-89 years participating in the Baltimore Longitudinal Study of Aging. Since excessive adiposity (body mass index ≥35 kg/m2 or waist-to-height ratio ≥0.62) may alter temperature set point, associations were also examined within adiposity strata. Overall, controlling for age, race, sex, height, exercise, and adiposity, lower temperature was associated with faster gait speed, less time to walk 400 m quickly, and lower perceived exertion following 5-min of walking at 0.67 m/s (all p ≤ 0.02). In the non-adipose (N = 662), these associations were more robust (all p ≤ 0.006). Direction of association was reversed in the adipose (N = 100), but none attained significance (all p > 0.22). Over 2.2 years, basal temperature was not associated with functional change in the overall population or non-adipose. Among the adipose, lower baseline temperature was associated with greater decline in endurance walking performance (p = 0.006). In longitudinal analyses predicting future functional performance, low temperature in the non-adipose was associated with faster gait speed (p = 0.021) and less time to walk 400 m quickly (p = 0.003), whereas in the adipose, lower temperature was associated with slower gait speed (p = 0.05) and more time to walk 400 m (p = 0.008). In older adults, lower basal body temperature appears to be associated with healthy aging in the absence of excessive adiposity.
Collapse
Affiliation(s)
- Eleanor M Simonsick
- Intramural Research Program National Institute on Aging, 3001 S. Hanover Street, 5th Floor, Baltimore, MD, 21225, USA.
| | - Helen C S Meier
- Joseph J. Zilber School of Public Health, University of Wisconsin-Madison, Wilwaukee, WI, USA
| | - Nancy Chiles Shaffer
- Intramural Research Program National Institute on Aging, 3001 S. Hanover Street, 5th Floor, Baltimore, MD, 21225, USA
| | - Stephanie A Studenski
- Intramural Research Program National Institute on Aging, 3001 S. Hanover Street, 5th Floor, Baltimore, MD, 21225, USA
| | - Luigi Ferrucci
- Intramural Research Program National Institute on Aging, 3001 S. Hanover Street, 5th Floor, Baltimore, MD, 21225, USA
| |
Collapse
|
280
|
Butsch WS, Stanford FC. Caloric restriction improves health-related quality of life in healthy normal weight and overweight individuals. ACTA ACUST UNITED AC 2016; 22:33. [PMID: 27881466 DOI: 10.1136/ebmed-2016-110526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- W Scott Butsch
- Harvard Medical School, Boston, Massachusetts, USA.,Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Fatima Cody Stanford
- Harvard Medical School, Boston, Massachusetts, USA.,Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
281
|
Rochon J, Bhapkar M, Pieper CF, Kraus WE. Application of the Marginal Structural Model to Account for Suboptimal Adherence in a Randomized Controlled Trial. Contemp Clin Trials Commun 2016; 4:222-228. [PMID: 27900372 PMCID: PMC5124349 DOI: 10.1016/j.conctc.2016.10.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Background There is considerable interest in adjusting for suboptimal adherence in randomized controlled trials. A per-protocol analysis, for example removes individuals who fail to achieve a minimal level of adherence. One can also reassign non-adherers to the control group, censor them at the point of non-adherence, or cross them over to the control. However, there are biases inherent in each of these methods. Here, we describe an application of causal modeling to address this issue. Methods The marginal structural model with inverse-probability weighting was implemented using a weighted generalized estimating equation model. Two ancillary models were developed to derive the weights. First, stepwise linear regression was used to model the observed percent weight loss, while stepwise logistic regression model was applied to model early discontinuation from the intervention. From these, participant- and time-specific weights were calculated. Discussion This model is complicated and requires careful attention to detail. Which variables to force into the ancillary models, how to construct interaction terms, and how to address time-dependent covariates must be considered. Nevertheless, it can be used to great effect to predict intervention effects at full adherence. Moreover, by contrasting these results against intention-to-treat results, insights can be gained into the intrinsic physiologic effect of the intervention. Trial registration ClinicalTrials.gov Identifier NCT00427193.
Collapse
Affiliation(s)
- James Rochon
- Rho Federal Systems, 6330 Quadrangle Drive, Chapel Hill, NC 27517, USA
| | - Manjushri Bhapkar
- Duke Clinical Research Institute, 2400 Pratt Street, Durham, NC 27710, USA
| | - Carl F Pieper
- Department of Biostatistics and Bioinformatics, Duke University Medical Center, 2424 Erwin Road, Durham, NC 27710, USA
| | - William E Kraus
- Duke Clinical Research Institute, 2400 Pratt Street, Durham, NC 27710, USA; Duke Molecular Physiology Institute, 300 North Duke Street, Durham, NC 27701, USA
| |
Collapse
|
282
|
Hesselink MKC, Schrauwen-Hinderling V, Schrauwen P. Skeletal muscle mitochondria as a target to prevent or treat type 2 diabetes mellitus. Nat Rev Endocrinol 2016; 12:633-645. [PMID: 27448057 DOI: 10.1038/nrendo.2016.104] [Citation(s) in RCA: 190] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Low levels of physical activity and the presence of obesity are associated with mitochondrial dysfunction. In addition, mitochondrial dysfunction has been associated with the development of insulin resistance and type 2 diabetes mellitus (T2DM). Although the evidence for a causal relationship between mitochondrial function and insulin resistance is still weak, emerging evidence indicates that boosting mitochondrial function might be beneficial to patient health. Exercise training is probably the most recognized promoter of mitochondrial function and insulin sensitivity and hence is still regarded as the best strategy to prevent and treat T2DM. Animal data, however, have revealed several new insights into the regulation of mitochondrial metabolism, and novel targets for interventions to boost mitochondrial function have emerged. Importantly, many of these targets seem to be regulated by factors such as nutrition, ambient temperature and circadian rhythms, which provides a basis for nonpharmacological strategies to prevent or treat T2DM in humans. Here, we will review the current evidence that mitochondrial function can be targeted therapeutically to improve insulin sensitivity and to prevent T2DM, focusing mainly on human intervention studies.
Collapse
Affiliation(s)
- Matthijs K C Hesselink
- Department of Human Biology and Human Movement Sciences, Maastricht University Medical Center, Universiteitsingel 50, 6229 ER, Maastricht, Netherlands
- NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Universiteitsingel 50, 6229 ER, Maastricht, Netherlands
| | - Vera Schrauwen-Hinderling
- Department of Human Biology and Human Movement Sciences, Maastricht University Medical Center, Universiteitsingel 50, 6229 ER, Maastricht, Netherlands
- NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Universiteitsingel 50, 6229 ER, Maastricht, Netherlands
- Department of Radiology, Maastricht University Medical Center, P. Debyelaan 25, 6229 HX, Maastricht, Netherlands
| | - Patrick Schrauwen
- Department of Human Biology and Human Movement Sciences, Maastricht University Medical Center, Universiteitsingel 50, 6229 ER, Maastricht, Netherlands
- NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Universiteitsingel 50, 6229 ER, Maastricht, Netherlands
| |
Collapse
|
283
|
Abstract
Despite billions of dollars spent over decades of research, debate remains over the causes and solutions of the obesity epidemic. The specific role of physical activity in the prevention or treatment of obesity seems a particularly contentious issue, with opposing views put forth in both academic and popular media. In an attempt to provide context and clarity to the specific question of the role of physical activity in determination of body weight, we have attempted to identify evidence or lack thereof in the scientific literature and provide a summary of our findings. Areas covered: Topics included in this narrative review are an overview of energy balance, the relationship between physical activity and energy expenditure, compensatory responses in non-exercise energy expenditure and energy intake, and the relationship between physical activity and obesity. Expert commentary: Based on a review of the existing literature, daily physical activity and structured exercise has beneficial effects on an individual's body weight. In most instances, exercise occurring in adequate amounts will increase total daily energy expenditure and create an acute energy deficit, without compensatory decreases in non-exercise physical activity or energy expenditure nor compensatory increases in energy intake. Several gaps in the literature exist, both in terms of the number of adequately powered clinical trials with rigorous assessments of both energy intake and expenditure, and with a variety of study populations (by age, sex, race, etc.) and with varying exercise volumes and intensities.
Collapse
Affiliation(s)
- Robin P Shook
- a Department of Kinesiology , Iowa State University , Ames , IA , USA
- b Department of Pediatrics , Children's Mercy Hospital , Kansas City , MO , USA
| |
Collapse
|
284
|
Serum concentrations and gene expression of sirtuin 1 in healthy and slightly overweight subjects after caloric restriction or resveratrol supplementation: A randomized trial. Int J Cardiol 2016; 227:788-794. [PMID: 28029409 DOI: 10.1016/j.ijcard.2016.10.058] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Revised: 10/17/2016] [Accepted: 10/22/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND Sirtuin 1 (Sirt1) plays an important role in vascular biology, and influences aspects of age-dependent atherosclerosis. In animals, the sirtuin system is strongly influenced by resveratrol and caloric restriction, but its expression in humans is controversial. This study investigated the effects of resveratrol and caloric restriction on Sirt1 serum concentrations and vascular biomarkers in a healthy human population. METHODS AND RESULTS Forty-eight healthy participants (24 women) aged 55-65years were randomized to either 30days of resveratrol administration (500mg/day) or caloric restriction (1000cal/day). Blood was collected at baseline and day 30. Laboratory data analyzed were triglycerides, total cholesterol, HDL, VLDL, LDL, apolipoprotein A1, apolipoprotein B, lipoprotein (a), non-esterified fatty acids (NEFA), glucose, insulin, oxidative stress, C-reactive protein, and Sirt1. Expression of the Sirt1 gene was analyzed using real-time PCR. Caloric restriction diminished the abdominal circumference and improved the lipid profile, but not resveratrol intervention. Resveratrol and caloric restriction increased serum concentrations of Sirt1, from 1.06±0.71 to 5.75±2.98ng/mL; p<0.0001, and from 1.65±1.81 to 5.80±2.23ng/mL; p<0.0001, respectively. Sirt1 increased in women and men in both interventions. On the other hand expression of Sirt1 mRNA was not different after caloric restriction and resveratrol treatment. CONCLUSIONS Caloric restriction and resveratrol significantly increased plasma concentrations of Sirt1. The long-term impact of these interventions on atherosclerosis should be assessed.
Collapse
|
285
|
Short-term caloric restriction in db/db mice improves myocardial function and increases high molecular weight (HMW) adiponectin. ACTA ACUST UNITED AC 2016; 13:28-34. [PMID: 27942464 DOI: 10.1016/j.ijcme.2016.10.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND Obesity and metabolic syndrome lead to the development of metabolic heart disease (MHD) that is characterized by left ventricular hypertrophy (LVH), diastolic dysfunction, and increased mitochondrial ROS. Caloric restriction (CR) is a nutritional intervention that protects against obesity, diabetes, and cardiovascular disease. Healthy adipose tissue is cardioprotective via releasing adipokines such as adiponectin. We tested the hypothesis that CR can ameliorate MHD and it is associated with improved adipose tissue function as reflected by increased circulating levels of high molecular weight (HMW) adiponectin and AMP-activated protein kinase (AMPK) in db/db mice. METHODS Genetically obese db/db and lean db/+ male mice were fed either ad libitum or subjected to 30% CR for 5 weeks. At the end of the study period, echocardiography was carried out to assess diastolic function. Blood, heart, and epididymal fat pads were harvested for mitochondrial study, ELISA, and Western blot analyses. RESULTS CR reversed the development of LVH, prevented diastolic dysfunction, and decreased cardiac mitochondrial H2O2 in db/db (vs. ad lib) mice. These beneficial effects on the heart were associated with increased circulating level of HMW adiponectin. Furthermore, CR increased AMPK and eNOS activation in white adipose tissue of db/db mice, but not in the heart. CONCLUSIONS These findings indicate that even short-term CR protects the heart from MHD. Whether the beneficial effects of CR on the heart could be related to the improved adipose tissue function warrants future investigation.
Collapse
|
286
|
Tam CS, Redman LM, Greenway F, LeBlanc KA, Haussmann MG, Ravussin E. Energy Metabolic Adaptation and Cardiometabolic Improvements One Year After Gastric Bypass, Sleeve Gastrectomy, and Gastric Band. J Clin Endocrinol Metab 2016; 101:3755-3764. [PMID: 27490919 PMCID: PMC6459006 DOI: 10.1210/jc.2016-1814] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
CONTEXT It is not known whether the magnitude of metabolic adaptation, a greater than expected drop in energy expenditure, depends on the type of bariatric surgery and is associated with cardiometabolic improvements. OBJECTIVE To compare changes in energy expenditure (metabolic chamber) and circulating cardiometabolic markers 8 weeks and 1 year after Roux-en-y bypass (RYGB), sleeve gastrectomy (SG), laparoscopic adjustable gastric band (LAGB), or a low-calorie diet (LCD). Design, Setting, Participants, and Intervention: This was a parallel-arm, prospective observational study of 30 individuals (27 females; mean age, 46 ± 2 years; body mass index, 47.2 ± 1.5 kg/m2) either self-selecting bariatric surgery (five RYGB, nine SG, seven LAGB) or on a LCD (n = 9) intervention (800 kcal/d for 8 weeks, followed by weight maintenance). RESULTS After 1 year, the RYGB and SG groups had similar degrees of body weight loss (33-36%), whereas the LAGB and LCD groups had 16 and 4% weight loss, respectively. After adjusting for changes in body composition, 24-hour energy expenditure was significantly decreased in all treatment groups at 8 weeks (-254 to -82 kcal/d), a drop that only persisted in RYGB (-124 ± 42 kcal/d; P = .002) and SG (-155 ± 118 kcal/d; P = .02) groups at 1 year. The degree of metabolic adaptation (24-hour and sleeping energy expenditure) was not significantly different between the treatment groups at either time-point. Plasma high-density lipoprotein and total and high molecular weight adiponectin were increased, and triglycerides and high-sensitivity C-reactive protein levels were reduced 1 year after RYGB or SG. CONCLUSIONS Metabolic adaptation of approximately 150 kcal/d occurs after RYGB and SG surgery. Future studies are required to examine whether these effects remain beyond 1 year.
Collapse
Affiliation(s)
- Charmaine S Tam
- Charles Perkins Centre and School of Life and Environmental Sciences (C.S.T.), The University of Sydney, Sydney, NSW 2006, Australia; Pennington Biomedical Research Center (C.S.T., L.M.R., F.G., E.R.), Baton Rouge, Louisiana 70808; and Our Lady of the Lake Physician Group (K.A.L., M.G.H.), Baton Rouge, Louisiana 70808
| | - Leanne M Redman
- Charles Perkins Centre and School of Life and Environmental Sciences (C.S.T.), The University of Sydney, Sydney, NSW 2006, Australia; Pennington Biomedical Research Center (C.S.T., L.M.R., F.G., E.R.), Baton Rouge, Louisiana 70808; and Our Lady of the Lake Physician Group (K.A.L., M.G.H.), Baton Rouge, Louisiana 70808
| | - Frank Greenway
- Charles Perkins Centre and School of Life and Environmental Sciences (C.S.T.), The University of Sydney, Sydney, NSW 2006, Australia; Pennington Biomedical Research Center (C.S.T., L.M.R., F.G., E.R.), Baton Rouge, Louisiana 70808; and Our Lady of the Lake Physician Group (K.A.L., M.G.H.), Baton Rouge, Louisiana 70808
| | - Karl A LeBlanc
- Charles Perkins Centre and School of Life and Environmental Sciences (C.S.T.), The University of Sydney, Sydney, NSW 2006, Australia; Pennington Biomedical Research Center (C.S.T., L.M.R., F.G., E.R.), Baton Rouge, Louisiana 70808; and Our Lady of the Lake Physician Group (K.A.L., M.G.H.), Baton Rouge, Louisiana 70808
| | - Mark G Haussmann
- Charles Perkins Centre and School of Life and Environmental Sciences (C.S.T.), The University of Sydney, Sydney, NSW 2006, Australia; Pennington Biomedical Research Center (C.S.T., L.M.R., F.G., E.R.), Baton Rouge, Louisiana 70808; and Our Lady of the Lake Physician Group (K.A.L., M.G.H.), Baton Rouge, Louisiana 70808
| | - Eric Ravussin
- Charles Perkins Centre and School of Life and Environmental Sciences (C.S.T.), The University of Sydney, Sydney, NSW 2006, Australia; Pennington Biomedical Research Center (C.S.T., L.M.R., F.G., E.R.), Baton Rouge, Louisiana 70808; and Our Lady of the Lake Physician Group (K.A.L., M.G.H.), Baton Rouge, Louisiana 70808
| |
Collapse
|
287
|
Abstract
Background Obesity is a consequence of chronic energy imbalance. We need accurate and precise measurements of energy intake and expenditure, as well as the related behaviors, to fully understand how energy homeostasis is regulated in order to develop interventions and evaluate their effectiveness to combat the global obesity epidemic. Scope of review We provide an in-depth review of the methodologies currently used to measure energy intake and expenditure in humans, including their principles, advantages, and limitations in the clinical research setting. The aim is to provide researchers with a comprehensive guide to conduct obesity research of the highest possible quality. Major conclusions An array of methodologies is available to measure various aspects of energy metabolism and none is perfect under all circumstances. The choice of methods should be specific to particular research questions with practicality and quality of data the priorities for consideration. A combination of complementary measurements may be preferable. There is an imperative need to develop new methodologies to improve the accuracy and precision of energy intake assessments. Image-based technology is a significant step to improve energy intake measurement. Physical activity informs patterns but not absolute energy expenditure. Combining complementary measurements overcomes shortfalls of individual methods.
Collapse
|
288
|
Eating disorders need more experimental psychopathology. Behav Res Ther 2016; 86:2-10. [PMID: 27600853 DOI: 10.1016/j.brat.2016.08.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2016] [Revised: 07/30/2016] [Accepted: 08/08/2016] [Indexed: 12/12/2022]
Abstract
Eating disorders are severe and disabling mental disorders. The scientific study of eating disorders has expanded dramatically over the past few decades, and provided significant understanding of eating disorders and their treatments. Those significant advances notwithstanding, there is scant knowledge about key processes that are crucial to clinical improvement. The lack of understanding mechanisms that cause, maintain and change eating disorders, currently is the biggest problem facing the science of eating disorders. It hampers the development of really effective interventions that could be fine-tuned to target the mechanisms of change and, therefore, the development of more effective treatments. It is argued here that the science of eating disorders and eating disorder treatment could benefit tremendously from pure experimental studies into its mechanisms of change, that is, experimental psychopathology (EPP). To illustrate why eating disorders need more EPP research, some key symptoms - restriction of intake, binge eating and body overvaluation - will be discussed. EPP studies challenge some generally accepted views and offer a fresh new look at key symptoms. This will, consequently, better inform eating disorder treatments.
Collapse
|
289
|
Cameron JL, Jain R, Rais M, White AE, Beer TM, Kievit P, Winters-Stone K, Messaoudi I, Varlamov O. Perpetuating effects of androgen deficiency on insulin resistance. Int J Obes (Lond) 2016; 40:1856-1863. [PMID: 27534842 PMCID: PMC5140744 DOI: 10.1038/ijo.2016.148] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 07/14/2016] [Accepted: 07/23/2016] [Indexed: 12/22/2022]
Abstract
Background/Objectives Androgen deprivation therapy (ADT) is commonly used for treatment of
prostate cancer, but is associated with side effects such as sarcopenia and
insulin resistance. The role of lifestyle factors such as diet and exercise
on insulin sensitivity and body composition in testosterone-deficient males
is poorly understood. The aim of the present study was to examine the
relationships between androgen status, diet, and insulin sensitivity. Subjects/Methods Middle-aged (11–12-yo) intact and orchidectomized male rhesus
macaques were maintained for two months on a standard chow diet, and then
exposed for six months to a Western-style, high-fat/calorie-dense diet (WSD)
followed by four months of caloric restriction (CR). Body composition,
insulin sensitivity, physical activity, serum cytokine levels, and adipose
biopsies were evaluated before and after each dietary intervention. Results Both intact and orchidectomized animals gained similar proportions of
body fat, developed visceral and subcutaneous adipocyte hypertrophy, and
became insulin resistant in response to the WSD. CR reduced body fat in both
groups, but reversed insulin resistance only in intact animals.
Orchidectomized animals displayed progressive sarcopenia, which persisted
after the switch to CR. Androgen deficiency was associated with increased
levels of interleukin-6 and macrophage-derived chemokine (CCL22), both of
which were elevated during CR. Physical activity levels showed a negative
correlation with body fat and insulin sensitivity. Conclusion Androgen deficiency exacerbated the negative metabolic side effects
of the WSD, such that CR alone was not sufficient to improve altered insulin
sensitivity, suggesting that ADT patients will require additional
interventions to reverse insulin resistance and sarcopenia.
Collapse
|
290
|
Tam CS, Rigas G, Heilbronn LK, Matisan T, Probst Y, Talbot M. Energy Adaptations Persist 2 Years After Sleeve Gastrectomy and Gastric Bypass. Obes Surg 2016; 26:459-63. [PMID: 26637359 DOI: 10.1007/s11695-015-1972-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Non-surgical weight loss induces a greater than expected decrease in energy expenditure, a phenomenon known as 'metabolic adaptation'. The effects of different bariatric surgery procedures on metabolic adaptation are not yet known and may partially contribute to weight loss success. We compared resting energy expenditure (REE) in 35 subjects (nine males; age = 46 ± 11 years; BMI = 42.1 ± 6.5 kg/m(2)) undergoing gastric band, sleeve gastrectomy or Roux-en-Y gastric bypass (RYGB) up to 2 years after surgery. We found a greater than expected reduction of 130-300 kcal/day at 6 weeks after sleeve and bypass surgery which was not explained by changes in body composition; this change was not seen in the band group. The suppression in REE after sleeve and RYGB remained up to 2 years, even after weight loss had plateaued. Our findings suggest that energy adaptation is not a contributing mechanism to medium-term weight maintenance after sleeve and RYGB bariatric surgeries.
Collapse
Affiliation(s)
- Charmaine S Tam
- The Charles Perkins Centre and School of Biological Sciences, University of Sydney, Sydney, Australia.
| | - Georgia Rigas
- Upper Gastrointestinal Unit, Department of Surgery, St George Private Hospital, Suite 3, Level 5, 1 South St, Kogarah, Australia.
| | - Leonie K Heilbronn
- Discipline of Medicine, University of Adelaide, Adelaide, South Australia, Australia.
| | - Tania Matisan
- Upper Gastrointestinal Unit, Department of Surgery, St George Private Hospital, Suite 3, Level 5, 1 South St, Kogarah, Australia.
| | - Yasmine Probst
- Smart Foods Centre, School of Medicine, University of Wollongong, Wollongong, NSW, Australia.
| | - Michael Talbot
- Upper Gastrointestinal Unit, Department of Surgery, St George Private Hospital, Suite 3, Level 5, 1 South St, Kogarah, Australia.
| |
Collapse
|
291
|
Frasca D, Diaz A, Romero M, Blomberg BB. Ageing and obesity similarly impair antibody responses. Clin Exp Immunol 2016; 187:64-70. [PMID: 27314456 DOI: 10.1111/cei.12824] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/08/2016] [Indexed: 12/19/2022] Open
Abstract
Ageing is characterized by increased low-grade chronic inflammation, which is a significant risk factor for morbidity and mortality of elderly individuals. Similar to ageing, obesity is considered to be an inflammatory predisposition associated with chronic activation of immune cells and consequent local and systemic inflammation. Both ageing and obesity are characterized by reduced innate and adaptive immune responses. This review focuses on B cells, how they may contribute, at least locally, to low-grade chronic inflammation in ageing and obesity and on the mechanisms involved.
Collapse
Affiliation(s)
- D Frasca
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, USA
| | - A Diaz
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, USA
| | - M Romero
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, USA
| | - B B Blomberg
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, USA
| |
Collapse
|
292
|
Teich T, Dunford EC, Porras DP, Pivovarov JA, Beaudry JL, Hunt H, Belanoff JK, Riddell MC. Glucocorticoid antagonism limits adiposity rebound and glucose intolerance in young male rats following the cessation of daily exercise and caloric restriction. Am J Physiol Endocrinol Metab 2016; 311:E56-68. [PMID: 27143556 PMCID: PMC4967147 DOI: 10.1152/ajpendo.00490.2015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 04/27/2016] [Indexed: 11/22/2022]
Abstract
Severe caloric restriction (CR), in a setting of regular physical exercise, may be a stress that sets the stage for adiposity rebound and insulin resistance when the food restriction and exercise stop. In this study, we examined the effect of mifepristone, a glucocorticoid (GC) receptor antagonist, on limiting adipose tissue mass gain and preserving whole body insulin sensitivity following the cessation of daily running and CR. We calorically restricted male Sprague-Dawley rats and provided access to voluntary running wheels for 3 wk followed by locking of the wheels and reintroduction to ad libitum feeding with or without mifepristone (80 mg·kg(-1)·day(-1)) for 1 wk. Cessation of daily running and CR increased HOMA-IR and visceral adipose mass as well as glucose and insulin area under the curve during an oral glucose tolerance test vs. pre-wheel lock exercised rats and sedentary rats (all P < 0.05). Insulin sensitivity and glucose tolerance were preserved and adipose tissue mass gain was attenuated by daily mifepristone treatment during the post-wheel lock period. These findings suggest that following regular exercise and CR there are GC-induced mechanisms that promote adipose tissue mass gain and impaired metabolic control in healthy organisms and that this phenomenon can be inhibited by the GC receptor antagonist mifepristone.
Collapse
Affiliation(s)
- Trevor Teich
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - Emily C Dunford
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - Deanna P Porras
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - Jacklyn A Pivovarov
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada
| | - Jacqueline L Beaudry
- Department of Medicine, Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada; and
| | - Hazel Hunt
- Corcept Therapeutics, Menlo Park, California
| | | | - Michael C Riddell
- Muscle Health Research Centre, School of Kinesiology and Health Science, York University, Toronto, Ontario, Canada;
| |
Collapse
|
293
|
Arabi YM, Al-Dorzi HM, McIntyre L, Mehta S. Design of nutrition trials in critically ill patients: food for thought. ANNALS OF TRANSLATIONAL MEDICINE 2016; 4:186. [PMID: 27275499 DOI: 10.21037/atm.2016.05.02] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Yaseen M Arabi
- 1 King Saud bin Abdulaziz University for Health Sciences and King Abdullah International Medical Research Center, Riyadh, Saudi Arabia ; 2 Department of Medicine, Division of Critical Care Medicine, University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada ; 3 Interdepartmental Division of Critical Care Medicine, Department of Medicine, Division of Respirology, University of Toronto, Mount Sinai Hospital, Toronto, Canada
| | - Hasan M Al-Dorzi
- 1 King Saud bin Abdulaziz University for Health Sciences and King Abdullah International Medical Research Center, Riyadh, Saudi Arabia ; 2 Department of Medicine, Division of Critical Care Medicine, University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada ; 3 Interdepartmental Division of Critical Care Medicine, Department of Medicine, Division of Respirology, University of Toronto, Mount Sinai Hospital, Toronto, Canada
| | - Lauralyn McIntyre
- 1 King Saud bin Abdulaziz University for Health Sciences and King Abdullah International Medical Research Center, Riyadh, Saudi Arabia ; 2 Department of Medicine, Division of Critical Care Medicine, University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada ; 3 Interdepartmental Division of Critical Care Medicine, Department of Medicine, Division of Respirology, University of Toronto, Mount Sinai Hospital, Toronto, Canada
| | - Sangeeta Mehta
- 1 King Saud bin Abdulaziz University for Health Sciences and King Abdullah International Medical Research Center, Riyadh, Saudi Arabia ; 2 Department of Medicine, Division of Critical Care Medicine, University of Ottawa, Ottawa Hospital Research Institute, Ottawa, Canada ; 3 Interdepartmental Division of Critical Care Medicine, Department of Medicine, Division of Respirology, University of Toronto, Mount Sinai Hospital, Toronto, Canada
| |
Collapse
|
294
|
Varlamov O. Western-style diet, sex steroids and metabolism. Biochim Biophys Acta Mol Basis Dis 2016; 1863:1147-1155. [PMID: 27264336 DOI: 10.1016/j.bbadis.2016.05.025] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Revised: 05/27/2016] [Accepted: 05/28/2016] [Indexed: 12/14/2022]
Abstract
The evolutionary transition from hunting to farming was associated with introduction of carbohydrate-rich diets. Today, the increased consumption of simple sugars and high-fat food brought about by Western-style diet and physical inactivity are leading causes of the growing obesity epidemic in the Western society. The extension of human lifespan far beyond reproductive age increased the burden of metabolic disorders associated with overnutrition and age-related hypogonadism. Sex steroids are essential regulators of both reproductive function and energy metabolism, whereas their imbalance causes infertility, obesity, glucose intolerance, dyslipidemia, and increased appetite. Clinical and translational studies suggest that dietary restriction and weight control can improve metabolic and reproductive outcomes of sex hormone-related pathologies, including testosterone deficiency in men and natural menopause and hyperandrogenemia in women. Minimizing metabolic and reproductive decline through rationally designed diet and exercise can help extend human reproductive age and promote healthy aging. This article is part of a Special Issue entitled: Oxidative Stress and Mitochondrial Quality in Diabetes/Obesity and Critical Illness Spectrum of Diseases - edited by P. Hemachandra Reddy.
Collapse
Affiliation(s)
- Oleg Varlamov
- Division of Diabetes, Obesity, and Metabolism, Oregon National Primate Research Center, Beaverton, OR 97006, United States.
| |
Collapse
|
295
|
Hume DJ, Yokum S, Stice E. Low energy intake plus low energy expenditure (low energy flux), not energy surfeit, predicts future body fat gain. Am J Clin Nutr 2016; 103:1389-96. [PMID: 27169833 PMCID: PMC4880998 DOI: 10.3945/ajcn.115.127753] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 03/29/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND There is a paucity of studies that have prospectively tested the energy surfeit theory of obesity with the use of objectively estimated energy intake and energy expenditure in humans. An alternative theory is that homeostatic regulation of body weight is more effective when energy intake and expenditure are both high (high energy flux), implying that low energy flux should predict weight gain. OBJECTIVE We aimed to examine the predictive relations of energy balance and energy flux to future weight gain and tested whether results were replicable in 2 independent samples. DESIGN Adolescents (n = 154) and college-aged women (n = 75) underwent 2-wk objective doubly labeled water, resting metabolic rate, and percentage of body fat measures at baseline. Percentage of body fat was measured annually for 3 y of follow-up for the adolescent sample and for 2 y of follow-up for the young adult sample. RESULTS Low energy flux, but not energy surfeit, predicted future increases in body fat in both studies. Furthermore, high energy flux appeared to prevent fat gain in part because it was associated with a higher resting metabolic rate. CONCLUSION Counter to the energy surfeit model of obesity, results suggest that increasing energy expenditure may be more effective for reducing body fat than caloric restriction, which is currently the treatment of choice for obesity. This trial was registered at clinicaltrials.gov as NCT02084836.
Collapse
Affiliation(s)
- David John Hume
- Division of Exercise Science and Sports Medicine, Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; and
| | | | | |
Collapse
|
296
|
Mitchell SE, Delville C, Konstantopedos P, Derous D, Green CL, Chen L, Han JDJ, Wang Y, Promislow DEL, Douglas A, Lusseau D, Speakman JR. The effects of graded levels of calorie restriction: III. Impact of short term calorie and protein restriction on mean daily body temperature and torpor use in the C57BL/6 mouse. Oncotarget 2016; 6:18314-37. [PMID: 26286956 PMCID: PMC4621893 DOI: 10.18632/oncotarget.4506] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Accepted: 07/13/2015] [Indexed: 11/30/2022] Open
Abstract
A commonly observed response in mammals to calorie restriction (CR) is reduced body temperature (Tb). We explored how the Tb of male C57BL/6 mice responded to graded CR (10 to 40%), compared to the response to equivalent levels of protein restriction (PR) over 3 months. Under CR there was a dynamic change in daily Tb over the first 30–35 days, which stabilized thereafter until day 70 after which a further decline was noted. The time to reach stability was dependent on restriction level. Body mass negatively correlated with Tb under ad libitum feeding and positively correlated under CR. The average Tb over the last 20 days was significantly related to the levels of body fat, structural tissue, leptin and insulin-like growth factor-1. Some mice, particularly those under higher levels of CR, showed periods of daily torpor later in the restriction period. None of the changes in Tb under CR were recapitulated by equivalent levels of PR. We conclude that changes in Tb under CR are a response only to the shortfall in calorie intake. The linear relationship between average Tb and the level of restriction supports the idea that Tb changes are an integral aspect of the lifespan effect.
Collapse
Affiliation(s)
- Sharon E Mitchell
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Camille Delville
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Penelope Konstantopedos
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Davina Derous
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Cara L Green
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - Luonan Chen
- Key laboratory of Systems Biology, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institute of Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jing-Dong J Han
- Chinese Academy of Sciences Key Laboratory of Computational Biology, Chinese Academy of Sciences-Max Planck Partner Institute for Computational Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yingchun Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Chaoyang, Beijing, China
| | - Daniel E L Promislow
- Department of Pathology, University of Washington at Seattle, Seattle, Washington, USA
| | - Alex Douglas
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - David Lusseau
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | - John R Speakman
- Institute of Biological and Environmental Sciences, University of Aberdeen, Aberdeen, Scotland, UK.,State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Chaoyang, Beijing, China
| |
Collapse
|
297
|
Alternating Diet as a Preventive and Therapeutic Intervention for High Fat Diet-induced Metabolic Disorder. Sci Rep 2016; 6:26325. [PMID: 27189661 PMCID: PMC4870701 DOI: 10.1038/srep26325] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 05/03/2016] [Indexed: 12/16/2022] Open
Abstract
This study presents the alternating diet as a new strategy in combating obesity and metabolic diseases. Lean or obese mice were fed a high-fat diet (HFD) for five days and switched to a regular diet for one (5 + 1), two (5 + 2), or five (5 + 5) days before switching back to HFD to start the second cycle, for a total of eight weeks (for prevention) or five weeks (for treatment) without limiting animals' access to food. Our results showed that animals with 5 + 2 and 5 + 5 diet alternations significantly inhibited body weight and fat mass gain compared to animals fed an HFD continuously. The dietary switch changed the pattern of daily caloric intake and suppressed HFD-induced adipose macrophage infiltration and chronic inflammation, resulting in improved insulin sensitivity and alleviated fatty liver. Alternating diet inhibited HFD-induced hepatic Pparγ-mediated lipid accumulation and activated the expression of Pparα and its target genes. Alternating diet in the 5 + 5 schedule induced weight loss in obese mice and reversed the progression of metabolic disorders, including hepatic steatosis, glucose intolerance, and inflammation. The results provide direct evidence to support that alternating diet represents a new intervention in dealing with the prevalence of diet-induced obesity.
Collapse
|
298
|
Venkatasubramanian S, Noh RM, Daga S, Langrish JP, Mills NL, Waterhouse BR, Hoffmann E, Jacobson EW, Lang NN, Frier BM, Newby DE. Effects of the small molecule SIRT1 activator, SRT2104 on arterial stiffness in otherwise healthy cigarette smokers and subjects with type 2 diabetes mellitus. Open Heart 2016; 3:e000402. [PMID: 27239324 PMCID: PMC4879341 DOI: 10.1136/openhrt-2016-000402] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 03/09/2016] [Accepted: 04/13/2016] [Indexed: 11/24/2022] Open
Abstract
Objective Arterial stiffness increases with age, and is associated with adverse cardiovascular outcome including increased mortality. The effect of the oral small molecule SIRT1 activator, SRT2104, on arterial stiffness was examined in otherwise healthy cigarette smokers and participants with type 2 diabetes mellitus. Methods 24 otherwise healthy cigarette smokers and 15 people with stable type 2 diabetes were randomised in a double-blind placebo-controlled crossover trial and received 28 days of oral SRT2104 (2.0 g/day) or matched placebo. Blood pressure was measured using non-invasive oscillatory sphygmomanometry. Pulse wave analysis and velocity were measured using applanation tonometry at baseline and the end of each treatment period. Owing to the small sample size and similar trends for both groups, data for the two groups were pooled (post hoc analysis). Results Compared to placebo, treatment with SRT2104 was associated with a significant reduction in augmentation pressure (p=0.0273) and a trend towards improvement in the augmentation index and corrected augmentation index (p>0.05 for both). However, no changes were observed in pulse wave velocity and time to wave reflection (p>0.05). Systolic and diastolic blood pressures remained unchanged throughout the study. Treatment by cohort interaction was not significant for any of the pulse wave parameters, suggesting that the response to SRT2104 in otherwise healthy smokers and people with diabetes was consistent. Conclusions SRT2104 may improve measures of arterial stiffness in otherwise healthy cigarette smokers and in participants with type 2 diabetes. Definitive conclusions are not possible given the small sample size and exploratory nature of this analysis. Trial registration number NCT01031108.
Collapse
Affiliation(s)
| | - Radzi M Noh
- Department of Diabetes , Royal Infirmary , Edinburgh , UK
| | | | - Jeremy P Langrish
- Centre for Cardiovascular Science, University of Edinburgh , Edinburgh , UK
| | - Nicholas L Mills
- Centre for Cardiovascular Science, University of Edinburgh , Edinburgh , UK
| | | | | | | | - Ninian N Lang
- Centre for Cardiovascular Science, University of Edinburgh , Edinburgh , UK
| | - Brian M Frier
- Department of Diabetes , Royal Infirmary , Edinburgh , UK
| | - David E Newby
- Centre for Cardiovascular Science, University of Edinburgh , Edinburgh , UK
| |
Collapse
|
299
|
Bankoglu EE, Seyfried F, Rotzinger L, Nordbeck A, Corteville C, Jurowich C, Germer CT, Otto C, Stopper H. Impact of weight loss induced by gastric bypass or caloric restriction on oxidative stress and genomic damage in obese Zucker rats. Free Radic Biol Med 2016; 94:208-17. [PMID: 26939878 DOI: 10.1016/j.freeradbiomed.2016.02.033] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 02/26/2016] [Accepted: 02/27/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND Evidence on bariatric surgery induced weight loss and its possible impact on cancer risk is limited, but also controversial. We used obese Zucker(fa/fa) and lean Zucker(fa/+) to investigate the association between obesity, oxidative stress and genomic damage after weight loss induced either by Roux-en-Y gastric bypass surgery (RYGB) or caloric restriction. METHODS Male Zucker(fa/fa) rats underwent RYGB (n=15) or sham surgery (n=17). Five shams were food restricted and body weight matched (BWM) to RYGB. Twelve Zucker(fa/+) rats served as lean controls. Body weight and food intake were measured daily. An oral glucose tolerance test was performed on day 27. DHE staining and western blots of HSP70 and HO-1 were used to evaluate oxidative stress and anti-3-nitrotyrosine antibody staining for nitrative stress detection in colon and kidney. Lipid peroxidation products in urine were quantified by TBARS assay. LC/MS/MS was applied to measure urinary excretion of 8-oxoGua (oxidized DNA derived base), 8-oxodG (oxidized DNA derived nucleoside) and 8-oxoGuo (oxidized RNA derived nucleoside). DNA double strand breaks (DSBs) and cell proliferation (PCNA) were detected by immunohistochemistry. RESULTS Sham-operated rats showed impaired glucose tolerance, elevated plasma insulin levels as well as elevated oxidative stress and nitrative stress markers, which were less severe after weight loss by RYGB or caloric restriction. Cell proliferation showed similar trends but no significant alteration. DNA DSBs were more frequent in sham-operated compared to all other groups. DNA damage in Zucker(fa/fa) rats positively correlated with basal plasma insulin values (Spearman's correlation coefficient for colon, 0.634 and for kidney, 0.525). CONCLUSIONS RYGB and caloric restriction were sufficient to significantly reduce elevated oxidative/nitrative stress and genomic damage in obese Zucker(fa/fa) rats. Further investigations are needed to elucidate the underlying mechanism of these genome protective effects.
Collapse
Affiliation(s)
- Ezgi Eyluel Bankoglu
- Institute of Pharmacology and Toxicology, University of Wuerzburg, Versbacher Str. 9, 97078 Würzburg, Germany
| | - Florian Seyfried
- Department of General, Visceral, Vascular and Paediatric Surgery, University Hospital of Wuerzburg, Germany
| | - Laura Rotzinger
- Department of General, Visceral, Vascular and Paediatric Surgery, University Hospital of Wuerzburg, Germany
| | - Arno Nordbeck
- Department of General, Visceral, Vascular and Paediatric Surgery, University Hospital of Wuerzburg, Germany
| | - Caroline Corteville
- Department of General, Visceral, Vascular and Paediatric Surgery, University Hospital of Wuerzburg, Germany
| | - Christian Jurowich
- Department of General, Visceral, Vascular and Paediatric Surgery, University Hospital of Wuerzburg, Germany
| | - Christoph Thomas Germer
- Department of General, Visceral, Vascular and Paediatric Surgery, University Hospital of Wuerzburg, Germany
| | - Christoph Otto
- Department of General, Visceral, Vascular and Paediatric Surgery, University Hospital of Wuerzburg, Germany; Experimental Surgery, Department of General, Visceral, Vascular, and Pediatric Surgery, University Hospital of Wuerzburg, Germany
| | - Helga Stopper
- Institute of Pharmacology and Toxicology, University of Wuerzburg, Versbacher Str. 9, 97078 Würzburg, Germany.
| |
Collapse
|
300
|
Morgan A, Mooney K, Wilkinson S, Pickles N, Mc Auley M. Cholesterol metabolism: A review of how ageing disrupts the biological mechanisms responsible for its regulation. Ageing Res Rev 2016; 27:108-124. [PMID: 27045039 DOI: 10.1016/j.arr.2016.03.008] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 03/22/2016] [Accepted: 03/30/2016] [Indexed: 02/06/2023]
Abstract
Cholesterol plays a vital role in the human body as a precursor of steroid hormones and bile acids, in addition to providing structure to cell membranes. Whole body cholesterol metabolism is maintained by a highly coordinated balancing act between cholesterol ingestion, synthesis, absorption, and excretion. The aim of this review is to discuss how ageing interacts with these processes. Firstly, we will present an overview of cholesterol metabolism. Following this, we discuss how the biological mechanisms which underpin cholesterol metabolism are effected by ageing. Included in this discussion are lipoprotein dynamics, cholesterol absorption/synthesis and the enterohepatic circulation/synthesis of bile acids. Moreover, we discuss the role of oxidative stress in the pathological progression of atherosclerosis and also discuss how cholesterol biosynthesis is effected by both the mammalian target of rapamycin and sirtuin pathways. Next, we examine how diet and alterations to the gut microbiome can be used to mitigate the impact ageing has on cholesterol metabolism. We conclude by discussing how mathematical models of cholesterol metabolism can be used to identify therapeutic interventions.
Collapse
|