1
|
Jacobsen JM, Petersen N, Torz L, Gerstenberg MK, Pedersen K, Østergaard S, Wulff BS, Andersen B, Raun K, Christoffersen BØ, John LM, Reitman ML, Kuhre RE. Housing mice near vs. below thermoneutrality affects drug-induced weight loss but does not improve prediction of efficacy in humans. Cell Rep 2024; 43:114501. [PMID: 39067024 PMCID: PMC11380917 DOI: 10.1016/j.celrep.2024.114501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/29/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024] Open
Abstract
Evaluation of weight loss drugs is usually performed in diet-induced obese mice housed at ∼22°C. This is a cold stress that increases energy expenditure by ∼35% compared to thermoneutrality (∼30°C), which may overestimate drug-induced weight loss. We investigated five anti-obesity mechanisms that have been in clinical development, comparing weight loss in mice housed at 22°C vs. 30°C. Glucagon-like peptide-1 (GLP-1), human fibroblast growth factor 21 (hFGF21), and melanocortin-4 receptor (MC4R) agonist induced similar weight losses. Peptide YY elicited greater vehicle-subtracted weight loss at 30°C (7.2% vs. 1.4%), whereas growth differentiation factor 15 (GDF15) was more effective at 22°C (13% vs. 6%). Independent of ambient temperature, GLP-1 and hFGF21 prevented the reduction in metabolic rate caused by weight loss. There was no simple rule for a better prediction of human drug efficacy based on ambient temperature, but since humans live at thermoneutrality, drug testing using mice should include experiments near thermoneutrality.
Collapse
Affiliation(s)
- Julie M Jacobsen
- Obesity and Liver Pharmacology, Integrated Physiology Research, Novo Nordisk A/S, Bagsværd, Denmark
| | - Natalia Petersen
- Liver and Gut Biology, Obesity & NASH, Global Drug Discovery, Novo Nordisk A/S, Bagsværd, Denmark
| | - Lola Torz
- Liver and Gut Biology, Obesity & NASH, Global Drug Discovery, Novo Nordisk A/S, Bagsværd, Denmark
| | | | - Kent Pedersen
- Obesity and Liver Pharmacology, Integrated Physiology Research, Novo Nordisk A/S, Bagsværd, Denmark
| | - Søren Østergaard
- Obesity and Liver Pharmacology, Integrated Physiology Research, Novo Nordisk A/S, Bagsværd, Denmark
| | - Birgitte S Wulff
- Obesity and Liver Pharmacology, Integrated Physiology Research, Novo Nordisk A/S, Bagsværd, Denmark
| | - Birgitte Andersen
- Diabetes, Obesity and NASH, Global Drug Discovery, Novo Nordisk A/S, Bagsværd, Denmark
| | - Kirsten Raun
- Lead Portfolio Projects, Research and Early Development, Novo Nordisk A/S, Bagsværd, Denmark
| | | | - Linu M John
- Obesity and Liver Pharmacology, Integrated Physiology Research, Novo Nordisk A/S, Bagsværd, Denmark
| | - Marc L Reitman
- Diabetes, Endocrinology, and Obesity Branch, National Institute of Diabetes and Digestive and Kidney Diseases, NIH, Bethesda, MD 20892, USA
| | - Rune E Kuhre
- Obesity and Liver Pharmacology, Integrated Physiology Research, Novo Nordisk A/S, Bagsværd, Denmark; Department of Biomedicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
2
|
Raun SH, Braun JL, Karavaeva I, Henriquez-Olguín C, Ali MS, Møller LLV, Gerhart-Hines Z, Fajardo VA, Richter EA, Sylow L. Mild Cold Stress at Ambient Temperature Elevates Muscle Calcium Cycling and Exercise Adaptations in Obese Female Mice. Endocrinology 2024; 165:bqae102. [PMID: 39136248 DOI: 10.1210/endocr/bqae102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Indexed: 08/28/2024]
Abstract
CONTEXT Housing temperature is a critical regulator of mouse metabolism and thermoneutral housing can improve model translation to humans. However, the impact of housing temperature on the ability of wheel running exercise training to rescue the detrimental effect of diet-induced obese mice is currently not fully understood. OBJECTIVE To investigate how housing temperature affects muscle metabolism in obese mice with regard to calcium handling and exercise training (ET) adaptations in skeletal muscle, and benefits of ET on adiposity and glucometabolic parameters. METHODS Lean or obese female mice were housed at standard ambient temperature (22 °C) or thermoneutrality (30 °C) with/without access to running wheels. The metabolic phenotype was investigated using glucose tolerance tests, indirect calorimetry, and body composition. Molecular muscle adaptations were measured using immunoblotting, qPCR, and spectrophotometric/fluorescent assays. RESULTS Obese female mice housed at 22 °C showed lower adiposity, lower circulating insulin levels, improved glucose tolerance, and elevated basal metabolic rate compared to 30 °C housing. Mice exposed to voluntary wheel running exhibited a larger fat loss and higher metabolic rate at 22 °C housing compared to thermoneutrality. In obese female mice, glucose tolerance improved after ET independent of housing temperature. Independent of diet and training, 22 °C housing increased skeletal muscle sarco(endo)plasmic reticulum Ca2+ ATPase (SERCA) activity. Additionally, housing at 22 °C elevated the induction of training-responsive muscle proteins in obese mice. CONCLUSION Our findings highlight that housing temperature significantly influences adiposity, insulin sensitivity, muscle physiology, and exercise adaptations in diet-induced obese female mice.
Collapse
Affiliation(s)
- Steffen H Raun
- Molecular Metabolism in Cancer and Ageing, Department of Biomedical Sciences, Faculty of Health, University of Copenhagen, Copenhagen 2200, Denmark
| | - Jessica L Braun
- Muscle Plasticity in Health and Disease, Department of Kinesiology, Faculty of Applied Health Sciences, Brock University, St. Catharines, L2A 3A1, Canada
| | - Iuliia Karavaeva
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health, University of Copenhagen, Copenhagen 2200, Denmark
| | - Carlos Henriquez-Olguín
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen 2100, Denmark
- Exercise Science Laboratory, Faculty of Medicine, Universidad Finis Terrae, Santiago 7501015, Chile
| | - Mona S Ali
- Molecular Metabolism in Cancer and Ageing, Department of Biomedical Sciences, Faculty of Health, University of Copenhagen, Copenhagen 2200, Denmark
| | - Lisbeth L V Møller
- Molecular Metabolism in Cancer and Ageing, Department of Biomedical Sciences, Faculty of Health, University of Copenhagen, Copenhagen 2200, Denmark
| | - Zachary Gerhart-Hines
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health, University of Copenhagen, Copenhagen 2200, Denmark
| | - Val A Fajardo
- Muscle Plasticity in Health and Disease, Department of Kinesiology, Faculty of Applied Health Sciences, Brock University, St. Catharines, L2A 3A1, Canada
| | - Erik A Richter
- The August Krogh Section for Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen 2100, Denmark
| | - Lykke Sylow
- Molecular Metabolism in Cancer and Ageing, Department of Biomedical Sciences, Faculty of Health, University of Copenhagen, Copenhagen 2200, Denmark
| |
Collapse
|
3
|
Winn NC, Schleh MW, Garcia JN, Lantier L, McGuinness OP, Blair JA, Hasty AH, Wasserman DH. Insulin at the intersection of thermoregulation and glucose homeostasis. Mol Metab 2024; 81:101901. [PMID: 38354854 PMCID: PMC10877958 DOI: 10.1016/j.molmet.2024.101901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/07/2024] [Accepted: 02/12/2024] [Indexed: 02/16/2024] Open
Abstract
Mammals are protected from changes in environmental temperature by altering energetic processes that modify heat production. Insulin is the dominant stimulus of glucose uptake and metabolism, which are fundamental for thermogenic processes. The purpose of this work was to determine the interaction of ambient temperature induced changes in energy expenditure (EE) on the insulin sensitivity of glucose fluxes. Short-term and adaptive responses to thermoneutral temperature (TN, ∼28 °C) and room (laboratory) temperature (RT, ∼22 °C) were studied in mice. This range of temperature does not cause detectable changes in circulating catecholamines or shivering and postabsorptive glucose homeostasis is maintained. We tested the hypothesis that a decrease in EE that occurs with TN causes insulin resistance and that this reduction in insulin action and EE is reversed upon short term (<12h) transition to RT. Insulin-stimulated glucose disposal (Rd) and tissue-specific glucose metabolic index were assessed combining isotopic tracers with hyperinsulinemic-euglycemic clamps. EE and insulin-stimulated Rd are both decreased (∼50%) in TN-adapted vs RT-adapted mice. When RT-adapted mice are switched to TN, EE rapidly decreases and Rd is reduced by ∼50%. TN-adapted mice switched to RT exhibit a rapid increase in EE, but whole-body insulin-stimulated Rd remains at the low rates of TN-adapted mice. In contrast, whole body glycolytic flux rose with EE. This higher EE occurs without increasing glucose uptake from the blood, but rather by diverting glucose from glucose storage to glycolysis. In addition to adaptations in insulin action, 'insulin-independent' glucose uptake in brown fat is exquisitely sensitive to thermoregulation. These results show that insulin action adjusts to non-stressful changes in ambient temperature to contribute to the support of body temperature homeostasis without compromising glucose homeostasis.
Collapse
Affiliation(s)
- Nathan C Winn
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
| | - Michael W Schleh
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Jamie N Garcia
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Louise Lantier
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; Vanderbilt Mouse Metabolic Phenotyping Center, Nashville, TN, USA
| | - Owen P McGuinness
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; Vanderbilt Mouse Metabolic Phenotyping Center, Nashville, TN, USA
| | - Joslin A Blair
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Alyssa H Hasty
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; VA Tennessee Valley Healthcare System, Nashville, TN, USA
| | - David H Wasserman
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; Vanderbilt Mouse Metabolic Phenotyping Center, Nashville, TN, USA
| |
Collapse
|
4
|
Zhu Y, Liu W, Qi Z. Adipose tissue browning and thermogenesis under physiologically energetic challenges: a remodelled thermogenic system. J Physiol 2024; 602:23-48. [PMID: 38019069 DOI: 10.1113/jp285269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 11/16/2023] [Indexed: 11/30/2023] Open
Abstract
Metabolic diseases such as obesity and diabetes are often thought to be caused by reduced energy expenditure, which poses a serious threat to human health. Cold exposure, exercise and caloric restriction have been shown to promote adipose tissue browning and thermogenesis. These physiological interventions increase energy expenditure and thus have emerged as promising strategies for mitigating metabolic disorders. However, that increased adipose tissue browning and thermogenesis elevate thermogenic consumption is not a reasonable explanation when humans and animals confront energetic challenges imposed by these interventions. In this review, we collected numerous results on adipose tissue browning and whitening and evaluated this bi-directional conversion of adipocytes from the perspective of energy homeostasis. Here, we propose a new interpretation of the role of adipose tissue browning under energetic challenges: increased adipose tissue browning and thermogenesis under energy challenge is not to enhance energy expenditure, but to reestablish a more economical thermogenic pattern to maintain the core body temperature. This can be achieved by enhancing the contribution of non-shivering thermogenesis (adipose tissue browning and thermogenesis) and lowering shivering thermogenesis and high intensity shivering. Consequently, the proportion of heat production in fat increases and that in skeletal muscle decreases, enabling skeletal muscle to devote more energy reserves to overcoming environmental stress.
Collapse
Affiliation(s)
- Yupeng Zhu
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention (Ministry of Education), East China Normal University, Shanghai, China
- School of Physical Education and Health, East China Normal University, Shanghai, China
- Sino-French Joint Research Center of Sport Science, East China Normal University, Shanghai, China
| | - Weina Liu
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention (Ministry of Education), East China Normal University, Shanghai, China
- School of Physical Education and Health, East China Normal University, Shanghai, China
| | - Zhengtang Qi
- The Key Laboratory of Adolescent Health Assessment and Exercise Intervention (Ministry of Education), East China Normal University, Shanghai, China
- School of Physical Education and Health, East China Normal University, Shanghai, China
| |
Collapse
|
5
|
Shirai T, Uemichi K, Takemasa T. Effects of the order of endurance and high-intensity interval exercise in combined training on mouse skeletal muscle metabolism. Am J Physiol Regul Integr Comp Physiol 2023; 325:R593-R603. [PMID: 37746708 DOI: 10.1152/ajpregu.00077.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/26/2023]
Abstract
Endurance exercise (EE) mainly improves oxidative capacity, whereas high-intensity interval exercise (HIIE) also improves glycolytic capacity. There is growing evidence that suggests that combining EE with HIIE can lead to improved athletic performance and fitness outcomes compared with either form of exercise alone. This study aimed to elucidate whether the order in which EE and HIIE are performed in combined training affects oxidative metabolism and glycolysis in mouse skeletal muscle. Male ICR mice at 7 wk of age were divided into three groups: control (CON), EE-HIIE, and HIIE-EE. The total training period was 3 wk (3 times/week). Mice performed running on a treadmill as endurance exercise and swimming with a weight load of 10% of body weight as high-intensity interval exercise. EE before HIIE (EE-HIIE) improved running performance in the maximal EE capacity test (all-out test) and partly enhanced the expression levels of molecular signals involved in glycolysis compared with HIIE before EE (HIIE-EE). The order of exercise did not, however, impact the expression of proteins related to mitochondrial dynamics, including those involved in the morphological changes of mitochondria through repeated fusion and fission, as well as oxidative energy metabolism. The findings suggest that the order of exercise has no significant impact on the expression of proteins associated with glycolytic and oxidative energy metabolism. Nevertheless, our results indicate that the order of EE-HIIE may enhance running performance.
Collapse
Affiliation(s)
- Takanaga Shirai
- Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Japan Society for Promotion Science, Chiyoda-ku, Tokyo, Japan
| | - Kazuki Uemichi
- Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Japan Society for Promotion Science, Chiyoda-ku, Tokyo, Japan
| | - Tohru Takemasa
- Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| |
Collapse
|
6
|
Townsend LK, Steinberg GR. AMPK and the Endocrine Control of Metabolism. Endocr Rev 2023; 44:910-933. [PMID: 37115289 DOI: 10.1210/endrev/bnad012] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/10/2023] [Accepted: 04/24/2023] [Indexed: 04/29/2023]
Abstract
Complex multicellular organisms require a coordinated response from multiple tissues to maintain whole-body homeostasis in the face of energetic stressors such as fasting, cold, and exercise. It is also essential that energy is stored efficiently with feeding and the chronic nutrient surplus that occurs with obesity. Mammals have adapted several endocrine signals that regulate metabolism in response to changes in nutrient availability and energy demand. These include hormones altered by fasting and refeeding including insulin, glucagon, glucagon-like peptide-1, catecholamines, ghrelin, and fibroblast growth factor 21; adipokines such as leptin and adiponectin; cell stress-induced cytokines like tumor necrosis factor alpha and growth differentiating factor 15, and lastly exerkines such as interleukin-6 and irisin. Over the last 2 decades, it has become apparent that many of these endocrine factors control metabolism by regulating the activity of the AMPK (adenosine monophosphate-activated protein kinase). AMPK is a master regulator of nutrient homeostasis, phosphorylating over 100 distinct substrates that are critical for controlling autophagy, carbohydrate, fatty acid, cholesterol, and protein metabolism. In this review, we discuss how AMPK integrates endocrine signals to maintain energy balance in response to diverse homeostatic challenges. We also present some considerations with respect to experimental design which should enhance reproducibility and the fidelity of the conclusions.
Collapse
Affiliation(s)
- Logan K Townsend
- Centre for Metabolism Obesity and Diabetes Research, Hamilton, ON L8S 4L8, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Gregory R Steinberg
- Centre for Metabolism Obesity and Diabetes Research, Hamilton, ON L8S 4L8, Canada
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, ON L8S 4L8, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
7
|
Pham T, Dollet L, Ali MS, Raun SH, Møller LL, Jafari A, Ditzel N, Andersen NR, Fritzen AM, Gerhart-Hines Z, Kiens B, Suomalainen A, Simpson SJ, Salling Olsen M, Kieser A, Schjerling P, Nieminen AI, Richter EA, Havula E, Sylow L. TNIK is a conserved regulator of glucose and lipid metabolism in obesity. SCIENCE ADVANCES 2023; 9:eadf7119. [PMID: 37556547 PMCID: PMC10411879 DOI: 10.1126/sciadv.adf7119] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 07/07/2023] [Indexed: 08/11/2023]
Abstract
Obesity and type 2 diabetes (T2D) are growing health challenges with unmet treatment needs. Traf2- and NCK-interacting protein kinase (TNIK) is a recently identified obesity- and T2D-associated gene with unknown functions. We show that TNIK governs lipid and glucose homeostasis in Drosophila and mice. Loss of the Drosophila ortholog of TNIK, misshapen, altered the metabolite profiles and impaired de novo lipogenesis in high sugar-fed larvae. Tnik knockout mice exhibited hyperlocomotor activity and were protected against diet-induced fat expansion, insulin resistance, and hepatic steatosis. The improved lipid profile of Tnik knockout mice was accompanied by enhanced skeletal muscle and adipose tissue insulin-stimulated glucose uptake and glucose and lipid handling. Using the T2D Knowledge Portal and the UK Biobank, we observed associations of TNIK variants with blood glucose, HbA1c, body mass index, body fat percentage, and feeding behavior. These results define an untapped paradigm of TNIK-controlled glucose and lipid metabolism.
Collapse
Affiliation(s)
- T. C. Phung Pham
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lucile Dollet
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Mona S. Ali
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Steffen H. Raun
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lisbeth L. V. Møller
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Abbas Jafari
- Department of Cellular and Molecular Medicine, University of Copenhagen, Copenhagen, Denmark
| | - Nicholas Ditzel
- Molecular Endocrinology and Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital and University of Southern Denmark, Odense, Denmark
- Biomedical Laboratory, The Faculty of Health Sciences, University of Southern Denmark, Denmark
| | - Nicoline R. Andersen
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Andreas M. Fritzen
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Zachary Gerhart-Hines
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Bente Kiens
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Anu Suomalainen
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Finland
- Helsinki University Hospital, HUS Diagnostic Center, Helsinki 00290, Finland
| | - Stephen J. Simpson
- Charles Perkins Centre, The University of Sydney, Camperdown 2006, Australia
- School of Life and Environmental Sciences, The University of Sydney, Camperdown, 2006, Australia
| | - Morten Salling Olsen
- Laboratory for Molecular Cardiology, Department of Cardiology, Copenhagen University Hospital, Rigshospitalet, Building 9312, Henrik Harpestrengs Vej 4C, Copenhagen 2100, Denmark
- Laboratory for Molecular Cardiology, Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Arnd Kieser
- Helmholtz Centre Munich–German Research Centre for Environmental Health, Research Unit Signaling and Translation, Ingolstaedter Landstr. 1, Neuherberg 85764, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Peter Schjerling
- Institute of Sports Medicine, Department of Orthopaedic Surgery M, Bispebjerg Hospital, Copenhagen, Denmark
- Center for Healthy Aging, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anni I. Nieminen
- FIMM Metabolomics Unit, Institute for Molecular Medicine Finland, University of Helsinki, Finland
| | - Erik A. Richter
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
| | - Essi Havula
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Finland
| | - Lykke Sylow
- Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Copenhagen, Denmark
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
8
|
Stroh AM, Stanford KI. Exercise-induced regulation of adipose tissue. Curr Opin Genet Dev 2023; 81:102058. [PMID: 37295241 PMCID: PMC10524364 DOI: 10.1016/j.gde.2023.102058] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 05/10/2023] [Accepted: 05/11/2023] [Indexed: 06/12/2023]
Abstract
Exercise induces various beneficial whole-body adaptations and can delay the onset of obesity, type 2 diabetes, and cardiovascular disease. While many of the beneficial effects of exercise on skeletal muscle and the cardiovascular system have been well established, recent studies have highlighted the role of exercise-induced improvements to adipose tissue that affect metabolic and whole-body health. Studies investigating exercise-induced adaptations of white adipose tissue (WAT) and brown adipose tissue (BAT) demonstrate modifications to glucose uptake, mitochondrial activity, and endocrine profile, and a beiging of WAT in rodents. This review discusses recent studies of the exercise-induced adaptations to WAT and BAT and their implications.
Collapse
Affiliation(s)
- Andrew M Stroh
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA. https://twitter.com/@AndrewStroh
| | - Kristin I Stanford
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA.
| |
Collapse
|
9
|
Wei W, Riley NM, Lyu X, Shen X, Guo J, Raun SH, Zhao M, Moya-Garzon MD, Basu H, Sheng-Hwa Tung A, Li VL, Huang W, Wiggenhorn AL, Svensson KJ, Snyder MP, Bertozzi CR, Long JZ. Organism-wide, cell-type-specific secretome mapping of exercise training in mice. Cell Metab 2023; 35:1261-1279.e11. [PMID: 37141889 PMCID: PMC10524249 DOI: 10.1016/j.cmet.2023.04.011] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 02/21/2023] [Accepted: 04/05/2023] [Indexed: 05/06/2023]
Abstract
There is a significant interest in identifying blood-borne factors that mediate tissue crosstalk and function as molecular effectors of physical activity. Although past studies have focused on an individual molecule or cell type, the organism-wide secretome response to physical activity has not been evaluated. Here, we use a cell-type-specific proteomic approach to generate a 21-cell-type, 10-tissue map of exercise training-regulated secretomes in mice. Our dataset identifies >200 exercise training-regulated cell-type-secreted protein pairs, the majority of which have not been previously reported. Pdgfra-cre-labeled secretomes were the most responsive to exercise training. Finally, we show anti-obesity, anti-diabetic, and exercise performance-enhancing activities for proteoforms of intracellular carboxylesterases whose secretion from the liver is induced by exercise training.
Collapse
Affiliation(s)
- Wei Wei
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Biology, Stanford University, Stanford, CA 94305, USA; Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Nicholas M Riley
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA; Department of Chemistry, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Xuchao Lyu
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA; Wu Tsai Human Performance Alliance, Stanford University, Stanford, CA 94305, USA
| | - Xiaotao Shen
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94035, USA
| | - Jing Guo
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Steffen H Raun
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Meng Zhao
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Maria Dolores Moya-Garzon
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Himanish Basu
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Alan Sheng-Hwa Tung
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA
| | - Veronica L Li
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA; Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Wentao Huang
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Amanda L Wiggenhorn
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA; Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Katrin J Svensson
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael P Snyder
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94035, USA; Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Carolyn R Bertozzi
- Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA; Department of Chemistry, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Jonathan Z Long
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Sarafan ChEM-H, Stanford University, Stanford, CA 94305, USA; Stanford Diabetes Research Center, Stanford University School of Medicine, Stanford, CA 94305, USA; Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Wu Tsai Human Performance Alliance, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
10
|
Hylander BL, Qiao G, Cortes Gomez E, Singh P, Repasky EA. Housing temperature plays a critical role in determining gut microbiome composition in research mice: Implications for experimental reproducibility. Biochimie 2023; 210:71-81. [PMID: 36693616 PMCID: PMC10953156 DOI: 10.1016/j.biochi.2023.01.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 01/03/2023] [Accepted: 01/20/2023] [Indexed: 01/22/2023]
Abstract
Preclinical mouse models are widely used for studying mechanisms of disease and responses to therapeutics, however there is concern about the lack of experimental reproducibility and failure to predict translational success. The gut microbiome has emerged as a regulator of metabolism and immunological processes in health and disease. The gut microbiome of mice differs by supplier and this affects experimental outcomes. We have previously reported that the mandated, mildly cool housing temperature for research mice (22°-26 °C) induces chronic adrenergic stress which suppresses anti-tumor immunity and promotes tumor growth compared to thermoneutral housing (30 °C). Therefore, we wondered how housing temperature affects the microbiome. Here, we demonstrate that the gut microbiome of BALB/c mice is easily modulated by a few degrees difference in temperature. Our results reveal significant differences between the gut microbiome of mice housed at 22°-23 °C vs. 30 °C. Although the genera vary, we consistently observed an enrichment of members of the family Lachnospiraceae when mice are housed at 22°-23 °C. These findings demonstrate that adrenergic stress and need for increased energy harvest to support thermogenesis, in addition to other factors such as diet, modulates the gut microbiome and this could be one mechanism by which housing temperature affects experimental outcomes. Additionally, tumor growth in mice housed at 30 °C also increases the proportion of Lachnospiraceae. The idea that stress can alter the gut microbiome and cause differences in experimental outcomes is applicable to mouse studies in general and is a variable that has significant potential to affect experimental reproducibility.
Collapse
Affiliation(s)
- Bonnie L Hylander
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton streets, Buffalo, NY, 14263, USA.
| | - Guanxi Qiao
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton streets, Buffalo, NY, 14263, USA.
| | - Eduardo Cortes Gomez
- Department of Biostatistics & Bioinformatics, Roswell Park Comprehensive Cancer Center, Elm & Carlton streets, Buffalo, NY, 14263, USA.
| | - Prashant Singh
- Genomics Shared Resource, Roswell Park Comprehensive Cancer Center, Elm & Carlton streets, Buffalo, NY, 14263, USA.
| | - Elizabeth A Repasky
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm & Carlton streets, Buffalo, NY, 14263, USA.
| |
Collapse
|
11
|
Nagai M, Moriyama M, Ishii C, Mori H, Watanabe H, Nakahara T, Yamada T, Ishikawa D, Ishikawa T, Hirayama A, Kimura I, Nagahara A, Naito T, Fukuda S, Ichinohe T. High body temperature increases gut microbiota-dependent host resistance to influenza A virus and SARS-CoV-2 infection. Nat Commun 2023; 14:3863. [PMID: 37391427 PMCID: PMC10313692 DOI: 10.1038/s41467-023-39569-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 06/20/2023] [Indexed: 07/02/2023] Open
Abstract
Fever is a common symptom of influenza and coronavirus disease 2019 (COVID-19), yet its physiological role in host resistance to viral infection remains less clear. Here, we demonstrate that exposure of mice to the high ambient temperature of 36 °C increases host resistance to viral pathogens including influenza virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). High heat-exposed mice increase basal body temperature over 38 °C to enable more bile acids production in a gut microbiota-dependent manner. The gut microbiota-derived deoxycholic acid (DCA) and its plasma membrane-bound receptor Takeda G-protein-coupled receptor 5 (TGR5) signaling increase host resistance to influenza virus infection by suppressing virus replication and neutrophil-dependent tissue damage. Furthermore, the DCA and its nuclear farnesoid X receptor (FXR) agonist protect Syrian hamsters from lethal SARS-CoV-2 infection. Moreover, we demonstrate that certain bile acids are reduced in the plasma of COVID-19 patients who develop moderate I/II disease compared with the minor severity of illness group. These findings implicate a mechanism by which virus-induced high fever increases host resistance to influenza virus and SARS-CoV-2 in a gut microbiota-dependent manner.
Collapse
Affiliation(s)
- Minami Nagai
- Division of Viral Infection, Department of Infectious Disease Control, International Research Center for Infectious Diseases, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Miyu Moriyama
- Division of Viral Infection, Department of Infectious Disease Control, International Research Center for Infectious Diseases, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Chiharu Ishii
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
| | - Hirotake Mori
- Department of General Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan
| | | | | | - Takuji Yamada
- Metagen Therapeutics, Inc., Yamagata, Japan
- Department of Life Science and Technology, Tokyo Institute of Technology, Tokyo, Japan
| | - Dai Ishikawa
- Metagen Therapeutics, Inc., Yamagata, Japan
- Laboratory for Regenerative Microbiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Gastroenterology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Takamasa Ishikawa
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
| | - Akiyoshi Hirayama
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan
| | - Ikuo Kimura
- Laboratory of Molecular Neurobiology, Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Department of Applied Biological Science, Graduate School of Agriculture, Tokyo University of Agriculture and Technology, Tokyo, Japan
| | - Akihito Nagahara
- Laboratory for Regenerative Microbiology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Department of Gastroenterology, Juntendo University Faculty of Medicine, Tokyo, Japan
| | - Toshio Naito
- Department of General Medicine, Juntendo University Faculty of Medicine, Tokyo, Japan.
| | - Shinji Fukuda
- Institute for Advanced Biosciences, Keio University, Yamagata, Japan.
- Metagen Therapeutics, Inc., Yamagata, Japan.
- Laboratory for Regenerative Microbiology, Juntendo University Graduate School of Medicine, Tokyo, Japan.
- Gut Environmental Design Group, Kanagawa Institute of Industrial Science and Technology, Kanagawa, Japan.
- Transborder Medical Research Center, University of Tsukuba, Ibaraki, Japan.
| | - Takeshi Ichinohe
- Division of Viral Infection, Department of Infectious Disease Control, International Research Center for Infectious Diseases, Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
12
|
Kesić M, Baković P, Farkaš V, Bagarić R, Kolarić D, Štefulj J, Čičin-Šain L. Constitutive Serotonin Tone as a Modulator of Brown Adipose Tissue Thermogenesis: A Rat Study. Life (Basel) 2023; 13:1436. [PMID: 37511811 PMCID: PMC10381595 DOI: 10.3390/life13071436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/21/2023] [Accepted: 06/22/2023] [Indexed: 07/30/2023] Open
Abstract
Brown adipose tissue (BAT), an important regulator of thermogenic and metabolic processes, is considered a promising target to combat metabolic disorders. The neurotransmitter and hormone serotonin (5HT) is a major modulator of energy homeostasis, with its central and peripheral pools acting in opposing ways. To better understand how individual variations in 5HT homeostasis influence the thermogenic functionality of BAT, we used a rat model consisting of two sublines with constitutively increased (high-5HT) or decreased (low-5HT) whole-body 5HT tone, developed by selective breeding for platelet 5HT parameters. We have shown that animals with constitutively low 5HT activity maintained at a standard housing temperature (22 °C) have greater interscapular BAT (iBAT) mass and higher iBAT metabolic activity (as evidenced by measurements of iBAT temperature and glucose uptake), accompanied by increased iBAT mRNA expression of key thermogenic genes, compared to animals with high 5HT tone. In response to further thermogenic challenges-intermittent cold exposure or treatment with a β3-adrenergic agonist-5HT sublines show several functional and molecular differences linking constitutively low endogenous 5HT tone to higher BAT activity/capacity. Overall, the results support a role of 5-HT in the control of BAT thermogenesis They also suggest that individuals with lower 5HT activity may be more sensitive to β3-adrenergic drugs.
Collapse
Affiliation(s)
- Maja Kesić
- Department of Molecular Biology, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia
| | - Petra Baković
- Department of Molecular Biology, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia
| | - Vladimir Farkaš
- Department of Experimental Physics, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia
| | - Robert Bagarić
- Department of Experimental Physics, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia
| | - Darko Kolarić
- Centre for Informatics and Computing, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia
| | - Jasminka Štefulj
- Department of Molecular Biology, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia
| | - Lipa Čičin-Šain
- Department of Molecular Biology, Ruđer Bošković Institute, HR-10000 Zagreb, Croatia
| |
Collapse
|
13
|
Félix-Soriano E, Sáinz N, Gil-Iturbe E, Castilla-Madrigal R, Celay J, Fernández-Galilea M, Pejenaute Á, Lostao MP, Martínez-Climent JA, Moreno-Aliaga MJ. Differential remodeling of subcutaneous white and interscapular brown adipose tissue by long-term exercise training in aged obese female mice. J Physiol Biochem 2023:10.1007/s13105-023-00964-2. [PMID: 37204588 DOI: 10.1007/s13105-023-00964-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 04/26/2023] [Indexed: 05/20/2023]
Abstract
Obesity exacerbates aging-induced adipose tissue dysfunction. This study aimed to investigate the effects of long-term exercise on inguinal white adipose tissue (iWAT) and interscapular brown adipose tissue (iBAT) of aged obese mice. Two-month-old female mice received a high-fat diet for 4 months. Then, six-month-old diet-induced obese animals were allocated to sedentarism (DIO) or to a long-term treadmill training (DIOEX) up to 18 months of age. In exercised mice, iWAT depot revealed more adaptability, with an increase in the expression of fatty acid oxidation genes (Cpt1a, Acox1), and an amelioration of the inflammatory status, with a favorable modulation of pro/antiinflammatory genes and lower macrophage infiltration. Additionally, iWAT of trained animals showed an increment in the expression of mitochondrial biogenesis (Pgc1a, Tfam, Nrf1), thermogenesis (Ucp1), and beige adipocytes genes (Cd137, Tbx1). In contrast, iBAT of aged obese mice was less responsive to exercise. Indeed, although an increase in functional brown adipocytes genes and proteins (Pgc1a, Prdm16 and UCP1) was observed, few changes were found on inflammation-related and fatty acid metabolism genes. The remodeling of iWAT and iBAT depots occurred along with an improvement in the HOMA index for insulin resistance and in glucose tolerance. In conclusion, long-term exercise effectively prevented the loss of iWAT and iBAT thermogenic properties during aging and obesity. In iWAT, the long-term exercise program also reduced the inflammatory status and stimulated a fat-oxidative gene profile. These exercise-induced adipose tissue adaptations could contribute to the beneficial effects on glucose homeostasis in aged obese mice.
Collapse
Affiliation(s)
- Elisa Félix-Soriano
- University of Navarra; Center for Nutrition Research and Department of Nutrition, Food Science and Physiology; School of Pharmacy and Nutrition, Pamplona, Spain
| | - Neira Sáinz
- University of Navarra; Center for Nutrition Research and Department of Nutrition, Food Science and Physiology; School of Pharmacy and Nutrition, Pamplona, Spain
| | - Eva Gil-Iturbe
- University of Navarra; Center for Nutrition Research and Department of Nutrition, Food Science and Physiology; School of Pharmacy and Nutrition, Pamplona, Spain
| | - Rosa Castilla-Madrigal
- University of Navarra; Center for Nutrition Research and Department of Nutrition, Food Science and Physiology; School of Pharmacy and Nutrition, Pamplona, Spain
| | - Jon Celay
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, University of Navarra, Pamplona, Spain
- CIBERONC, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Marta Fernández-Galilea
- University of Navarra; Center for Nutrition Research and Department of Nutrition, Food Science and Physiology; School of Pharmacy and Nutrition, Pamplona, Spain
| | - Álvaro Pejenaute
- University of Navarra; Center for Nutrition Research and Department of Nutrition, Food Science and Physiology; School of Pharmacy and Nutrition, Pamplona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN). Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - M Pilar Lostao
- University of Navarra; Center for Nutrition Research and Department of Nutrition, Food Science and Physiology; School of Pharmacy and Nutrition, Pamplona, Spain
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN). Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - José A Martínez-Climent
- Division of Hemato-Oncology, Center for Applied Medical Research CIMA, University of Navarra, Pamplona, Spain
- CIBERONC, Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain
| | - María J Moreno-Aliaga
- University of Navarra; Center for Nutrition Research and Department of Nutrition, Food Science and Physiology; School of Pharmacy and Nutrition, Pamplona, Spain.
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN). Instituto de Salud Carlos III (ISCIII), Madrid, Spain.
- IdiSNA, Navarra Institute for Health Research, Pamplona, Spain.
| |
Collapse
|
14
|
Dreher SI, Irmler M, Pivovarova-Ramich O, Kessler K, Jürchott K, Sticht C, Fritsche L, Schneeweiss P, Machann J, Pfeiffer AFH, Hrabě de Angelis M, Beckers J, Birkenfeld AL, Peter A, Niess AM, Weigert C, Moller A. Acute and long-term exercise adaptation of adipose tissue and skeletal muscle in humans: a matched transcriptomics approach after 8-week training-intervention. Int J Obes (Lond) 2023; 47:313-324. [PMID: 36774413 PMCID: PMC10113153 DOI: 10.1038/s41366-023-01271-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 01/31/2023] [Accepted: 01/31/2023] [Indexed: 02/13/2023]
Abstract
BACKGROUND Exercise exerts many health benefits by directly inducing molecular alterations in physically utilized skeletal muscle. Molecular adaptations of subcutaneous adipose tissue (SCAT) might also contribute to the prevention of metabolic diseases. AIM To characterize the response of human SCAT based on changes in transcripts and mitochondrial respiration to acute and repeated bouts of exercise in comparison to skeletal muscle. METHODS Sedentary participants (27 ± 4 yrs) with overweight or obesity underwent 8-week supervised endurance exercise 3×1h/week at 80% VO2peak. Before, 60 min after the first and last exercise bout and 5 days post intervention, biopsies were taken for transcriptomic analyses and high-resolution respirometry (n = 14, 8 female/6 male). RESULTS In SCAT, we found 37 acutely regulated transcripts (FC > 1.2, FDR < 10%) after the first exercise bout compared to 394, respectively, in skeletal muscle. Regulation of only 5 transcripts overlapped between tissues highlighting their differential response. Upstream and enrichment analyses revealed reduced transcripts of lipid uptake, storage and lipogenesis directly after exercise in SCAT and point to β-adrenergic regulation as potential major driver. The data also suggest an exercise-induced modulation of the circadian clock in SCAT. Neither term was associated with transcriptomic changes in skeletal muscle. No evidence for beigeing/browning was found in SCAT along with unchanged respiration. CONCLUSIONS Adipose tissue responds completely distinct from adaptations of skeletal muscle to exercise. The acute and repeated reduction in transcripts of lipid storage and lipogenesis, interconnected with a modulated circadian rhythm, can counteract metabolic syndrome progression toward diabetes.
Collapse
Affiliation(s)
- Simon I Dreher
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, 72076, Tübingen, Germany
| | - Martin Irmler
- Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
| | - Olga Pivovarova-Ramich
- German Center for Diabetes Research (DZD), 85784, Neuherberg, Germany
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Potsdam, Germany
- Research Group Molecular Nutritional Medicine, German Institute of Human Nutrition Potsdam-Rehbruecke, 14558, Nuthetal, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology, Diabetes and Nutrition, Campus Benjamin Franklin, 12203, Berlin, Germany
| | - Katharina Kessler
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Potsdam, Germany
| | - Karsten Jürchott
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), 13353, Berlin, Germany
| | - Carsten Sticht
- Next Generation Sequencing Core Facility, Medical Faculty Mannheim, Heidelberg University, 68167, Mannheim, Germany
| | - Louise Fritsche
- German Center for Diabetes Research (DZD), 85784, Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München, University of Tübingen, Tübingen, Germany
| | - Patrick Schneeweiss
- Sports Medicine, University Hospital Tübingen, 72076, Tübingen, Germany
- Interfaculty Research Institute for Sport and Physical Activity, University of Tübingen, Tübingen, Germany
| | - Jürgen Machann
- German Center for Diabetes Research (DZD), 85784, Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München, University of Tübingen, Tübingen, Germany
- Section on Experimental Radiology, Department of Diagnostic and Interventional Radiology, University Hospital Tübingen, 72076, Tübingen, Germany
| | - Andreas F H Pfeiffer
- German Center for Diabetes Research (DZD), 85784, Neuherberg, Germany
- Department of Clinical Nutrition, German Institute of Human Nutrition Potsdam-Rehbruecke (DIfE), 14558, Nuthetal, Potsdam, Germany
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Department of Endocrinology, Diabetes and Nutrition, Campus Benjamin Franklin, 12203, Berlin, Germany
| | - Martin Hrabě de Angelis
- Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85784, Neuherberg, Germany
- Chair of Experimental Genetics, Technical University of Munich, 85354, Freising, Germany
| | - Johannes Beckers
- Institute of Experimental Genetics, Helmholtz Zentrum München, Ingolstädter Landstr. 1, 85764, Neuherberg, Germany
- German Center for Diabetes Research (DZD), 85784, Neuherberg, Germany
- Chair of Experimental Genetics, Technical University of Munich, 85354, Freising, Germany
| | - Andreas L Birkenfeld
- German Center for Diabetes Research (DZD), 85784, Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München, University of Tübingen, Tübingen, Germany
- Department of Internal Medicine IV, University Hospital Tübingen, 72076, Tübingen, Germany
| | - Andreas Peter
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, 72076, Tübingen, Germany
- German Center for Diabetes Research (DZD), 85784, Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München, University of Tübingen, Tübingen, Germany
| | - Andreas M Niess
- Sports Medicine, University Hospital Tübingen, 72076, Tübingen, Germany
- Interfaculty Research Institute for Sport and Physical Activity, University of Tübingen, Tübingen, Germany
| | - Cora Weigert
- Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital Tübingen, 72076, Tübingen, Germany.
- German Center for Diabetes Research (DZD), 85784, Neuherberg, Germany.
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München, University of Tübingen, Tübingen, Germany.
| | - Anja Moller
- German Center for Diabetes Research (DZD), 85784, Neuherberg, Germany
- Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Zentrum München, University of Tübingen, Tübingen, Germany
- Department of Internal Medicine IV, University Hospital Tübingen, 72076, Tübingen, Germany
| |
Collapse
|
15
|
Desai M, Torsoni AS, Torsoni MA, Eisaghalian A, Ferrini M, Ross MG. Thermoneutrality effects on developmental programming of obesity. J Dev Orig Health Dis 2023; 14:223-230. [PMID: 36097652 PMCID: PMC9998331 DOI: 10.1017/s2040174422000502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Developmental programming studies using mouse models have housed the animals at human thermoneutral temperatures (22°C) which imposes constant cold stress. As this impacts energy homeostasis, we investigated the effects of two housing temperatures (22°C and 30°C) on obesity development in male and female offspring of Control and FR dams. Pregnant mice were housed at 22°C (cold-exposed, CE) or 30°C (thermoneutrality, TN) room temperature. At gestational age e10, mice were fed either an ad libitum diet (Control) or were 30% food-restricted (FR) to produce low birth weight newborns. Following delivery, all dams were fed an ad libitum diet and maternal mice continued to nurse their own pups. At 3 weeks of age, offspring were weaned to an ad libitum diet and housed at similar temperatures as their mothers. Body weights and food intake were monitored. At 6 months of age, body composition and glucose tolerance test were determined, after which, brain and adipose tissue were collected for analysis. FR/CE and FR/TN offspring exhibited hyperphagia and were significantly heavier with increased adiposity as compared to their respective Controls. There was sex-specific effects of temperature in both groups. Male offspring at TN were heavier with increased body fat, though the food intake was decreased as compared to CE males. This was reflected by hypertrophic adipocytes and increased arcuate nucleus satiety/appetite ratio. In contrast, female offspring were not impacted by housing temperature. Thus, unlike female offspring, there was a significant interaction of diet and temperature evident in the male offspring with accentuated adverse effects evident in FR/TN males.
Collapse
Affiliation(s)
- Mina Desai
- Perinatal Research Laboratory, The Lundquist Institute at Harbor-UCLA Medical Center, Department of Obstetrics and Gynecology, Torrance, CA
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA
| | - Adrianna S. Torsoni
- Laboratory of Metabolic Disorders (Labdime), Faculty of Applied Sciences (FCA) of the University of Campinas (UNICAMP), Limeira/SP, Brazil
| | - Marcio A Torsoni
- Laboratory of Metabolic Disorders (Labdime), Faculty of Applied Sciences (FCA) of the University of Campinas (UNICAMP), Limeira/SP, Brazil
| | | | - Monica Ferrini
- Charles R. Drew University of Medicine and Science, Los Angeles, CA
| | - Michael G. Ross
- Perinatal Research Laboratory, The Lundquist Institute at Harbor-UCLA Medical Center, Department of Obstetrics and Gynecology, Torrance, CA
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA
- Department of Obstetrics and Gynecology, Charles R. Drew University, Los Angeles, CA
| |
Collapse
|
16
|
Hu Y, Zheng Y, Wang Y, Yu S, Li M, Zhou J, Wu G, Xing W, Dong L, Liu Y, Fang G, Li J, Zhao J, Zhang X, Gao F. Temperature-dependent metabolite orchestration to acute submaximal exercise indicates cardiorespiratory fitness in humans. Life Sci 2023; 313:121284. [PMID: 36529280 DOI: 10.1016/j.lfs.2022.121284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
AIMS Cardiorespiratory fitness (CRF), an important biomarker of human health, is impaired in cold environment compared to thermoneutral condition. The study aimed to investigate the role of metabolome response to acute exercise in regulation of CRF at different ambient temperatures. MAIN METHODS A total of 27 young adults were recruited, and each subject underwent a cardiopulmonary exercise test (CPET) and a constant load submaximal exercise at both room temperature (25 °C) and cold temperature (0 °C). The serum samples were collected before and immediately after constant load exercise. KEY FINDINGS Acute cold exposure decreased CRF by 41 %, accompanied by a metabolic shift to anaerobic respiration. It also decreased VO2 and increased respiratory quotient during constant load exercise. Metabolome profiling revealed that acute exercise reprogrammed serum metabolome in an ambient temperature-dependent manner. Specifically, exercise increased a cluster of fatty acids during cold exposure, possibly due to impaired fatty acid oxidation. The correlations between metabolite responses to acute exercise and exercise parameters were analyzed using partial least squares regression and machine learning, revealing that metabolite responses to acute exercise were highly correlated with exercise parameters and predictive of CRF. Among the contributors, tryptophan and its metabolites stood out as important ones. SIGNIFICANCE These results suggested that the metabolite responses to acute submaximal exercise unmasks the exercise performance at different ambient temperatures, highlighting the role of metabolite orchestration in the physiological regulation of CRF.
Collapse
Affiliation(s)
- Yang Hu
- Key Laboratory of Aerospace Medicine of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Yao Zheng
- School of Biomedical Engineering, Fourth Military Medical University, Xi'an 710032, China
| | - Yongchun Wang
- Key Laboratory of Aerospace Medicine of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Sen Yu
- Key Laboratory of Aerospace Medicine of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Min Li
- Key Laboratory of Aerospace Medicine of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Jiaheng Zhou
- Key Laboratory of Aerospace Medicine of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Guiling Wu
- Key Laboratory of Aerospace Medicine of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Wenjuan Xing
- Key Laboratory of Aerospace Medicine of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Ling Dong
- Key Laboratory of Aerospace Medicine of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Yang Liu
- School of Biomedical Engineering, Fourth Military Medical University, Xi'an 710032, China
| | - Guoliang Fang
- Exercise Biological Center, China Institute of Sport Science, Beijing 100061, China
| | - Jia Li
- Key Laboratory of Aerospace Medicine of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Jiexiu Zhao
- Exercise Biological Center, China Institute of Sport Science, Beijing 100061, China
| | - Xing Zhang
- Key Laboratory of Aerospace Medicine of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China.
| | - Feng Gao
- Key Laboratory of Aerospace Medicine of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an 710032, China.
| |
Collapse
|
17
|
Zhang G, Wang Y, Li R, Peng J, Zhang J, Hu R, Zhang L, Wu Y, Sun Q, Liu C. Sex difference in effects of intermittent heat exposure on hepatic lipid and glucose metabolisms. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 854:158704. [PMID: 36108838 DOI: 10.1016/j.scitotenv.2022.158704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/18/2022] [Accepted: 09/08/2022] [Indexed: 06/15/2023]
Abstract
Global climate warming has drawn worldwide attention. However, the health impact of heat exposure is still controversial. This study aimed to explore the exact effects and sex differential vulnerability under intermittent heat exposure (IHE) patterns and tried to elucidate the potential mechanisms by which IHE modulated hepatic lipid and glucose homeostasis. Both female and male C57BL/6 N mice were randomly allocated to control group (22 ± 1 °C) or intermittent heat group (37 ± 1 °C for 6 h) for 9 consecutive days followed by 4-day recovery at 22 ± 1 °C in a whole-body exposure chamber. Male mice, but not female, being influenced by IHE with decreased body weight, improved insulin sensitivity and glucose tolerance. Next, the levels of hepatic triglyceride (TG) were decreased and free fatty acid (FFA) increased in male mice exposed to intermittent heat, accompanied with upregulated expression of anti-oxidative enzymes in the liver. In addition, IHE led to enhanced lipid catabolism in male mice by inducing fatty acid uptake, lipid lipolysis, mitochondrial/peroxisomal fatty acid oxidation and lipid export. And glycolysis and glucose utilization were induced by IHE in male mice as well. Mechanically, heat shock protein 70 (HSP70)/insulin receptor substrate 1 (IRS1)/AMPKα pathways were activated in response to IHE. These findings provide new evidence that IHE sex-dependently enhanced the metabolism of lipid and glucose in male mice through HSP70/IRS1/AMPKα signaling.
Collapse
Affiliation(s)
- Guoqing Zhang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, Zhejiang, China
| | - Yindan Wang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, Zhejiang, China
| | - Ran Li
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, Zhejiang, China
| | - Jing Peng
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, Zhejiang, China
| | - Jinna Zhang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, Zhejiang, China
| | - Renjie Hu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, Zhejiang, China
| | - Lu Zhang
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, Zhejiang, China
| | - Yunlu Wu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, Zhejiang, China
| | - Qinghua Sun
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, Zhejiang, China
| | - Cuiqing Liu
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China; Zhejiang International Science and Technology Cooperation Base of Air Pollution and Health, Hangzhou, Zhejiang, China.
| |
Collapse
|
18
|
Unger CA, Hope MC, Aladhami AK, Velázquez KT, Enos RT. How stable is your vivarium's temperature? Fluctuations in vivarium temperature significantly impact metabolism and behavior impeding scientific reproducibility. Physiol Behav 2023; 258:114029. [PMID: 36372225 PMCID: PMC10797230 DOI: 10.1016/j.physbeh.2022.114029] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 11/07/2022] [Accepted: 11/08/2022] [Indexed: 11/12/2022]
Abstract
OBJECTIVES The purpose of this investigation was to examine the variability in vivarium temperature and the impact that this has on metabolic and behavioral outcomes in mice. METHODS Daily vivarium temperature was monitored every day for a two-year period. Behavioral and metabolic phenotyping were assessed in male and female C57BL/6 (n = 71/sex) mice over the course of 2 years. RESULTS Vivarium temperature was found to fluctuate on a monthly, daily, and even an hourly basis of approximately ±5ºC. A 5ºC change in temperature was found to result in daily changes in total energy expenditure (35% and 27%), resting energy expenditure (39% for both sexes), movement (51% and 37%), food consumption (35% and 29%), and sleep duration (15% and 13%) for female and male mice, respectively. CONCLUSIONS Fluctuations in vivarium temperature can dramatically impact metabolic and behavioral outcomes, which impedes scientific reproducibility. This awareness and the guidelines we propose in this publication will hopefully help to enhance the reproducibility of pre-clinical animal research.
Collapse
Affiliation(s)
- Christian A Unger
- Department of Pathology, Microbiology, and Immunology, University of South Carolina-School of Medicine, Columbia, SC, United States
| | - Marion C Hope
- Department of Pathology, Microbiology, and Immunology, University of South Carolina-School of Medicine, Columbia, SC, United States
| | - Ahmed K Aladhami
- Department of Pathology, Microbiology, and Immunology, University of South Carolina-School of Medicine, Columbia, SC, United States; University of Baghdad, Nursing College, Baghdad, Iraq
| | - Kandy T Velázquez
- Department of Pathology, Microbiology, and Immunology, University of South Carolina-School of Medicine, Columbia, SC, United States
| | - Reilly T Enos
- Department of Pathology, Microbiology, and Immunology, University of South Carolina-School of Medicine, Columbia, SC, United States.
| |
Collapse
|
19
|
Daneshyar S, Ghasemnian A, Mirakhori Z, Daneshyar S. The effect of high fat diet and endurance training on newly discovery of nonshivering-thermogenic factors under thermoneutrality in mice. Sci Sports 2022. [DOI: 10.1016/j.scispo.2022.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
20
|
Sadler DG, Treas L, Sikes JD, Porter C. A modest change in housing temperature alters whole body energy expenditure and adipocyte thermogenic capacity in mice. Am J Physiol Endocrinol Metab 2022; 323:E517-E528. [PMID: 36351253 PMCID: PMC9744648 DOI: 10.1152/ajpendo.00079.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 10/28/2022] [Accepted: 11/04/2022] [Indexed: 11/10/2022]
Abstract
Typical vivarium temperatures (20-26°C) induce facultative thermogenesis in mice, a process attributable in part to uncoupling protein-1 (UCP1). The impact of modest changes in housing temperature on whole body and adipose tissue energetics in mice remains unclear. Here, we determined the effects of transitioning mice from 24°C to 30°C on total energy expenditure and adipose tissue protein signatures. C57BL/6J mice were housed at 24°C for 2 wk and then either remained at 24°C (n = 16/group, 8M/8F) or were transitioned to 30°C (n = 16/group, 8M/8F) for 4 wk. Total energy expenditure and its components were determined by indirect calorimetry. Interscapular brown adipose tissue (iBAT) and inguinal white adipose tissue (iWAT) proteins were quantified by Western blot and quantitative proteomics. Transitioning from 24°C to 30°C reduced total energy expenditure in both male (-25%) and female (-16%) mice, which was attributable to lower basal energy expenditure in males (-36%) and females (-40%). Total iBAT UCP1 protein content was 50% lower at 30°C compared with 24°C, whereas iWAT UCP1 protein content was similar between conditions. iBAT UCP1 protein content remained 20-fold greater than iWAT at 30°C. In iBAT and iWAT, 183 and 41 proteins were differentially expressed between 24°C and 30°C, respectively. iWAT proteins (257) differentially expressed between sexes at 30°C were not differentially expressed at 24°C. Thus, 30°C housing lowers total energy expenditure of mice when compared with an ambient temperature (24°C) that falls within the National Research Council's guidelines for housing laboratory mice. Lower iBAT UCP1 content accompanied chronic housing at 30°C. Furthermore, housing temperature influences sexual dimorphism in the iWAT proteome. These data have implications regarding the optimization of preclinical models of human disease.NEW & NOTEWORTHY Housing mice at 30°C reduced the basal and total energy expenditure compared with 24°C, which was accompanied by a reduction in brown adipose tissue UCP1 content. Proteomic profiling demonstrated the brown adipose tissue and white adipose tissue proteomes were largely influenced by housing temperature and sex, respectively. Therefore, 30°C housing revealed sexual dimorphism in the white adipose tissue proteome that was largely absent in animals housed at 24°C.
Collapse
Affiliation(s)
- Daniel G Sadler
- Arkansas Children's Research Institute, Arkansas Children's Nutrition Center and the University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Lillie Treas
- Arkansas Children's Research Institute, Arkansas Children's Nutrition Center and the University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - James D Sikes
- Arkansas Children's Research Institute, Arkansas Children's Nutrition Center and the University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Craig Porter
- Arkansas Children's Research Institute, Arkansas Children's Nutrition Center and the University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
21
|
Weber A, Medak KD, Townsend LK, Wright DC. Ketogenic diet induced weight loss occurs independent of housing temperature and is followed by hyperphagia and weight regain after cessation in mice. J Physiol 2022; 600:4677-4693. [PMID: 36083198 DOI: 10.1113/jp283469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/24/2022] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Ketogenic diets reduce food intake, increase energy expenditure and cause weight loss in rodents Prior preclinical studies have been completed at room temperature, a condition which induces thermal stress and limits clinical translatability We demonstrate that ketogenic diet-induced reductions in food intake, increases in energy expenditure, weight loss and improvements in glucose homeostasis are similar in mice housed at room temperature or thermal neutrality Ketogenic diet induced reductions in food intake appear to explain a large degree of weight loss. Similarly, switching mice from a ketogenic to an obesogenic diet leads to hyperphagia mediated weight gain ABSTRACT: Ketogenic diets (KDs) are a popular tool used for weight management. Studies in mice have demonstrated that KDs reduce food intake, increase energy expenditure and cause weight loss. These studies were completed at room temperature (RT), a condition below the animal's thermal neutral (TN) zone which induces thermal stress. As energy intake and expenditure are sensitive to environmental temperature it's not clear if a KD would exert the same beneficial effects under TN conditions. Adherence to restrictive diets is poor and consequently it is important to examine the effects, and underlying mechanisms, of cycling from a ketogenic to an obesogenic diet. The purpose of the current study was to determine if housing temperature impacted the effects of a KD in obese mice and to determine if the mechanisms driving KD-induced weight loss reverse when mice are switched to an obesogenic high fat diet. We demonstrate that KD-induced reductions in food intake, increases in energy expenditure, weight loss and improvements in glucose homeostasis are not dependent upon housing temperature. KD-induced weight loss, seems to be largely explained by reductions in caloric intake while cycling mice back to an obesogenic diet following a period of KD feeding leads to hyperphagia-induced weight gain. Collectively, our results suggest that prior findings with mice fed a KD at RT are likely not an artifact of how mice were housed and that initial changes in weight when transitioning from an obesogenic to a ketogenic diet or back, are largely dependent on food intake. Abstract figure legend The impact of housing temperature on ketogenic diet mediated changes in energy expenditure, food intake and weight gain. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Alyssa Weber
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Kyle D Medak
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Logan K Townsend
- Centre for Metabolism, Obesity and Diabetes Research, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - David C Wright
- School of Kinesiology, University of British Columbia, Vancouver, British Columbia, Canada.,Faculty of Land and Food Systems, University of British Columbia, Vancouver, British Columbia, Canada.,British Columbia Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| |
Collapse
|
22
|
Mušo M, Bentley L, Vizor L, Yon M, Burling K, Barker P, Zolkiewski LAK, Cox RD, Dumbell R. A Wars2 mutant mouse shows a sex and diet specific change in fat distribution, reduced food intake and depot-specific upregulation of WAT browning. Front Physiol 2022; 13:953199. [PMID: 36091365 PMCID: PMC9452902 DOI: 10.3389/fphys.2022.953199] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 07/19/2022] [Indexed: 11/21/2022] Open
Abstract
Background: Increased waist-to-hip ratio (WHR) is associated with increased mortality and risk of type 2 diabetes and cardiovascular disease. The TBX15-WARS2 locus has consistently been associated with increased WHR. Previous study of the hypomorphic Wars2 V117L/V117L mouse model found phenotypes including severely reduced fat mass, and white adipose tissue (WAT) browning, suggesting Wars2 could be a potential modulator of fat distribution and WAT browning. Methods: To test for differences in browning induction across different adipose depots of Wars2 V117L/V117L mice, we measured multiple browning markers of a 4-month old chow-fed cohort in subcutaneous and visceral WAT and brown adipose tissue (BAT). To explain previously observed fat mass loss, we also tested for the upregulation of plasma mitokines FGF21 and GDF15 and for differences in food intake in the same cohort. Finally, to test for diet-associated differences in fat distribution, we placed Wars2 V117L/V117L mice on low-fat or high-fat diet (LFD, HFD) and assessed their body composition by Echo-MRI and compared terminal adipose depot weights at 6 months of age. Results: The chow-fed Wars2 V117L/V117L mice showed more changes in WAT browning marker gene expression in the subcutaneous inguinal WAT depot (iWAT) than in the visceral gonadal WAT depot (gWAT). These mice also demonstrated reduced food intake and elevated plasma FGF21 and GDF15, and mRNA from heart and BAT. When exposed to HFD, the Wars2 V117L/V117L mice showed resistance to diet-induced obesity and a male and HFD-specific reduction of gWAT: iWAT ratio. Conclusion: Severe reduction of Wars2 gene function causes a systemic phenotype which leads to upregulation of FGF21 and GDF15, resulting in reduced food intake and depot-specific changes in browning and fat mass.
Collapse
Affiliation(s)
- Milan Mušo
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, United Kingdom
| | - Liz Bentley
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, United Kingdom
- Mary Lyon Centre at MRC Harwell, Oxfordshire, United Kingdom
| | - Lucie Vizor
- Mary Lyon Centre at MRC Harwell, Oxfordshire, United Kingdom
| | - Marianne Yon
- Mary Lyon Centre at MRC Harwell, Oxfordshire, United Kingdom
| | - Keith Burling
- MRC Metabolic Diseases Unit, Mouse Biochemistry Laboratory, Cambridge, United Kingdom
| | - Peter Barker
- MRC Metabolic Diseases Unit, Mouse Biochemistry Laboratory, Cambridge, United Kingdom
| | | | - Roger D. Cox
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, United Kingdom
| | - Rebecca Dumbell
- Mammalian Genetics Unit, MRC Harwell Institute, Oxfordshire, United Kingdom
- Department of Biosciences, School of Science and Technology, Nottingham Trent University, Nottingham, United Kingdom
| |
Collapse
|
23
|
Brunetta HS, Townsend LK. Muscle-fat crosstalk: effects of exercise on brown adipose tissue, what do we know? J Physiol 2022; 600:4039-4040. [PMID: 35866569 DOI: 10.1113/jp283516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 06/30/2022] [Indexed: 11/08/2022] Open
|
24
|
Wang L, Zhou B, Li X, Wang Y, Yang XM, Wang H, Yan J, Dong J. The beneficial effects of exercise on glucose and lipid metabolism during statin therapy is partially mediated by changes of the intestinal flora. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2022; 41:112-120. [PMID: 35854693 PMCID: PMC9246419 DOI: 10.12938/bmfh.2021-024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 02/03/2022] [Indexed: 11/09/2022]
Abstract
Recent research has confirmed that moderate-intensity exercise affects the gut microbiome
composition and improves cardiac function in an animal model after myocardial infarction
(MI). However, few studies have investigated the effects of exercise on glucose and lipid
metabolism in patients with coronary heart disease (CHD) receiving a statin treatment and
successful percutaneous coronary intervention (PCI). Meanwhile, since statin therapy may
lead to the risk of an increase in blood glucose level in CHD patients, we hypothesized
that moderate-intensity exercise may be helpful for regulating glucose-lipid metabolism
and stabilizing the blood glucose level in CHD patients. Therefore, to confirm our
conjecture, we conducted a clinical retrospective study and animal experiment,
respectively. The clinical study involved a total of 501 statin-treated patients with CHD
after PCI. According to the study protocol, patients were divided into the following three
groups: a non-exercise group, exercise at the recommended standard group, and exercise not
at the recommended standard group. We found that qualified moderate-intensity exercise
decreased blood glucose and lipid levels at follow-up at a mean of 2.2 years, and the
incidence of new-onset diabetes showed a downward trend compared with the non-exercise and
exercise not at the recommended standard groups. Furthermore, we used a high-fat rat model
to explore an additional mechanism of the beneficial effects of exercise-based management
on glucose-lipid metabolism apart from the known mechanism. We used 16S rRNA
high-throughput sequencing technology to analyze the changes induced by exercise in the
composition of intestinal flora in experimental rats. We found that rats that exercised
with or without statin administration had lower plasma glucose and lipid levels and that
these parameters were higher in the control and statin-treated rats that did not exercise.
These results were consistent with the human study. The results from high-throughput
sequencing of the intestinal flora of rats showed, to the best of our knowledge, that
exercise leads to an increased relative abundance of Akkermansia
muciniphila, which contributes to improved glucose and lipid metabolism. Based
on our current results, we suggest that moderate-intensity exercise can improve glucose
and lipid metabolism and prevent statin treatment-related side effects, such as
hyperglycemia, in patients after PCI. Exercise could facilitate the applicability of
statins for lower lipid levels. Exercise training also provides additional benefits, such
as alteration of the gut microbiota, which contributes to improved glucose and lipid
metabolism.
Collapse
Affiliation(s)
- Lijun Wang
- Department of Cardiology, Affiliated Zhongshan Hospital, Dalian University, Dalian 116001, China.,Department of Cardiology, Affiliated Zhongshan Hospital, Dalian University, Dalian 116001, China
| | - Baihua Zhou
- Department of Cardiology, Affiliated Zhongshan Hospital, Dalian University, Dalian 116001, China.,Department of Respiratory and Critical Care Medicine, Yueyang Second People's Hospital, Yueyang, Hunan, China
| | - Xinying Li
- Department of Cardiology, Affiliated Zhongshan Hospital, Dalian University, Dalian 116001, China
| | - Yang Wang
- Department of Cardiology, First Affiliated Hospital of Medical School, Xi'an Jiaotong University, Xi'an 710061, China
| | - Xiu Mei Yang
- Department of Cardiology, Affiliated Zhongshan Hospital, Dalian University, Dalian 116001, China
| | - Hongwei Wang
- Department of Cardiology, Affiliated Zhongshan Hospital, Dalian University, Dalian 116001, China
| | - Jun Yan
- Department of Cardiology, Affiliated Zhongshan Hospital, Dalian University, Dalian 116001, China
| | - Jiakun Dong
- Department of Cardiology, Affiliated Zhongshan Hospital, Dalian University, Dalian 116001, China
| |
Collapse
|
25
|
Kim HJ, Kim YJ, Seong JK. AMP-activated protein kinase activation in skeletal muscle modulates exercise-induced uncoupled protein 1 expression in brown adipocyte in mouse model. J Physiol 2022; 600:2359-2376. [PMID: 35301717 PMCID: PMC9322297 DOI: 10.1113/jp282999] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/04/2022] [Indexed: 12/02/2022] Open
Abstract
Abstract Aerobic exercise is an effective intervention in preventing obesity and is also an important factor associated with thermogenesis. There is an increasing interest in the factors and mechanisms induced by aerobic exercise that can influence the metabolism and thermogenic activity in an individual. Recent studies suggest that exercise induced circulating factors (known as ‘exerkines’), which are able to modulate activation of brown adipose tissue (BAT) and browning of white adipose tissue. However, the underlying molecular mechanisms associated with the effect of exercise‐induced peripheral factors on BAT activation remain poorly understood. Furthermore, the role of exercise training in BAT activation is still debatable. Hence, the purpose of our study is to assess whether exercise training affects the expression of uncoupled protein 1 (UCP1) in brown adipocytes via release of different blood factors. Four weeks of exercise training significantly decreased the body weight gain and fat mass gain. Furthermore, trained mice exhibit higher levels of energy expenditure and UCP1 expression than untrained mice. Surprisingly, treatment with serum from exercise‐trained mice increased the expression of UCP1 in differentiated brown adipocytes. To gain a better understanding of these mechanisms, we analysed the conditioned media obtained after treating the C2C12 myotubes with an AMP‐activated protein kinase (AMPK) activator (AICAR; 5‐aminoimidazole‐4‐carboxamide ribonucleotide), which leads to an increased expression of UCP1 when added to brown adipocytes. Our observations suggest the possibility of aerobic exercise‐induced BAT activation via activation of AMPK in skeletal muscles. Key points Exercise promotes thermogenesis by activating uncoupling protein 1 (UCP1), which leads to a decrease in the body weight gain and body fat content. However, little is known about the role of exerkines in modulating UCP1 expression and subsequent brown adipose tissue (BAT) activation. Four weeks of voluntary wheel‐running exercise reduces body weight and fat content. Exercise induces the increase in AMP‐activated protein kinase (AMPK) and slow‐type muscle fibre marker genes in skeletal muscles and promotes UCP1 expression in white and brown adipose tissues. Incubation of brown adipocytes with serum isolated from exercise‐trained mice significantly increased their UCP1 gene and protein levels; moreover, conditioned media of AMPK‐activator‐treated C2C12 myotubes induces increased UCP1 expression in brown adipocytes. These results show that aerobic exercise‐induced skeletal muscle AMPK has a significant effect on UCP1 expression in BAT.
Collapse
Affiliation(s)
- Hye Jin Kim
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea.,Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, 08826, Republic of Korea
| | - Youn Ju Kim
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea.,Laboratory of Developmental Biology and Genomics, BK21 Program for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea.,Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, 08826, Republic of Korea
| | - Je Kyung Seong
- The Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, 08826, Republic of Korea.,Laboratory of Developmental Biology and Genomics, BK21 Program for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, South Korea.,Korea Mouse Phenotyping Center (KMPC), Seoul National University, Seoul, 08826, Republic of Korea.,Interdisciplinary Program for Bioinformatics, Program for Cancer Biology, BIO-MAX/N-Bio Institute, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
26
|
Tanimura R, Kobayashi L, Shirai T, Takemasa T. Effects of exercise intensity on white adipose tissue browning and its regulatory signals in mice. Physiol Rep 2022; 10:e15205. [PMID: 35286020 PMCID: PMC8919700 DOI: 10.14814/phy2.15205] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/21/2022] [Accepted: 01/21/2022] [Indexed: 05/10/2023] Open
Abstract
Adipose tissue has been classified into white adipose tissue (WAT), brown adipose tissue (BAT), and beige adipose tissue the latter of which is produced as WAT changes into BAT due to exposure to cold temperature or exercise. In response to these stimulations, WAT produces heat by increasing mitochondrial contents and the expression of uncoupling protein 1 (UCP1), thus facilitating browning. Exercise is known to be one of the triggers for WAT browning, but the effects of exercise intensity on the browning of WAT remain to be unclear. Therefore, in this study, we aimed to examine the effects of high- or low-intensity exercises on the browning of WAT. Mice performed high- or low-intensity running on a treadmill running 3 days a week for four weeks. As per our findings, it was determined that four weeks of running did not significantly reduce inguinal WAT (iWAT) wet weight but did significantly reduce adipocytes size, regardless of exercise intensity. The protein expression level of UCP1 was significantly increased in iWAT by high-intensity running. In addition, the expression of oxidative phosphorylation proteins (OXPHOS) in iWAT was significantly increased by high-intensity running. These results demonstrated that high-intensity exercise might be effective for increasing mitochondrial contents and heat production capacity in iWAT. Furthermore, we found that high-intensity running increased the protein expression level of fibroblast growth factor 21 (FGF21) in skeletal muscle compared with that in low intensity running. We have also examined the relationship between browning of WAT and the expression of FGF21 in skeletal muscle and found a positive correlation between the protein expression of UCP1 in iWAT and the protein expression of FGF21 in gastrocnemius muscle. In conclusion, we suggest that high-intensity exercise is effective for the browning of WAT and the increase of FGF21 in skeletal muscle.
Collapse
Affiliation(s)
- Riku Tanimura
- Graduate School of Comprehensive Human SciencesUniversity of TsukubaTsukubaJapan
| | - Leo Kobayashi
- Graduate School of Comprehensive Human SciencesUniversity of TsukubaTsukubaJapan
- JIJI PRESS LtdCyuo‐kuJapan
| | - Takanaga Shirai
- Research Fellow of the Japan Society for the Promotion of ScienceTokyoJapan
- Faculty of Health and Sports SciencesUniversity of TsukubaTsukubaJapan
| | - Tohru Takemasa
- Faculty of Health and Sports SciencesUniversity of TsukubaTsukubaJapan
| |
Collapse
|
27
|
Cold Exposure Drives Weight Gain and Adiposity following Chronic Suppression of Brown Adipose Tissue. Int J Mol Sci 2022; 23:ijms23031869. [PMID: 35163791 PMCID: PMC8836787 DOI: 10.3390/ijms23031869] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/17/2022] Open
Abstract
Therapeutic activation of thermogenic brown adipose tissue (BAT) may be feasible to prevent, or treat, cardiometabolic disease. However, rodents are commonly housed below thermoneutrality (~20 °C) which can modulate their metabolism and physiology including the hyperactivation of brown (BAT) and beige white adipose tissue. We housed animals at thermoneutrality from weaning to chronically supress BAT, mimic human physiology and explore the efficacy of chronic, mild cold exposure (20 °C) and β3-adrenoreceptor agonism (YM-178) under these conditions. Using metabolic phenotyping and exploratory proteomics we show that transfer from 28 °C to 20 °C drives weight gain and a 125% increase in subcutaneous fat mass, an effect not seen with YM-178 administration, thus suggesting a direct effect of a cool ambient temperature in promoting weight gain and further adiposity in obese rats. Following chronic suppression of BAT, uncoupling protein 1 mRNA was undetectable in the subcutaneous inguinal white adipose tissue (IWAT) in all groups. Using exploratory adipose tissue proteomics, we reveal novel gene ontology terms associated with cold-induced weight gain in BAT and IWAT whilst Reactome pathway analysis highlights the regulation of mitotic (i.e., G2/M transition) and metabolism of amino acids and derivatives pathways. Conversely, YM-178 had minimal metabolic-related effects but modified pathways involved in proteolysis (i.e., eukaryotic translation initiation) and RNA surveillance across both tissues. Taken together these findings are indicative of a novel mechanism whereby animals increase body weight and fat mass following chronic suppression of adaptive thermogenesis from weaning. In addition, treatment with a B3-adrenoreceptor agonist did not improve metabolic health in obese animals raised at thermoneutrality.
Collapse
|
28
|
Chen X, Bollinger E, Cunio T, Damilano F, Stansfield JC, Pinkus CA, Kreuser S, Hirenallur-Shanthappa DK, Roth Flach RJ. An assessment of thermoneutral housing conditions on murine cardiometabolic function. Am J Physiol Heart Circ Physiol 2021; 322:H234-H245. [PMID: 34919456 DOI: 10.1152/ajpheart.00461.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mouse models are used to model human diseases and perform pharmacological efficacy testing to advance therapies to humans; most of these studies are conducted in room temperature conditions. At room temperature (22°C), mice are cold stressed and must utilize brown adipose tissue (BAT) to maintain body temperature. This cold stress increases catecholamine tone to maintain adipocyte lipid release via lipolysis, which will fuel adaptive thermogenesis. Maintaining rodents at thermoneutral temperatures (28°C) ameliorates the need for adaptive thermogenesis, thus reducing catecholamine tone and BAT activity. Cardiovascular tone is also determined by catecholamine levels in rodents, as beta adrenergic stimuli are primary drivers of not only lipolytic, but also ionotropic and chronotropic responses. As mice have increased catecholamine tone at room temperature, we investigated how thermoneutral housing conditions would impact cardiometabolic function. Here, we show a rapid and reversible effect of thermoneutrality on both heart rate and blood pressure in chow fed animals, which was blunted in animals fed high fat diet. Animals subjected to transverse aortic constriction displayed compensated hypertrophy at room temperature, while animals displayed less hypertrophy and trends towards worse systolic function at thermoneutrality. Despite these dramatic changes in blood pressure and heart rate at thermoneutral housing conditions, enalapril effectively improved cardiac hypertrophy and gene expression alterations. There were surprisingly few differences in cardiac parameters in high fat fed animals at thermoneutrality. Overall, these data suggest that thermoneutral housing may alter some aspects of cardiac remodeling in preclinical mouse models of heart failure.
Collapse
Affiliation(s)
- Xian Chen
- Comparative Medicine, Pfizer Inc. Cambridge MA, United States
| | - Eliza Bollinger
- Internal Medicine Research Unit, Pfizer Inc. Cambridge MA, United States
| | - Teresa Cunio
- Comparative Medicine, Pfizer Inc. Cambridge MA, United States
| | - Federico Damilano
- Internal Medicine Research Unit, Pfizer Inc. Cambridge MA, United States
| | | | - Cynthia A Pinkus
- Internal Medicine Research Unit, Pfizer Inc. Cambridge MA, United States
| | - Steven Kreuser
- Comparative Medicine, Pfizer Inc. Cambridge MA, United States
| | | | | |
Collapse
|
29
|
Macêdo APA, Muñoz VR, Cintra DE, Pauli JR. 12,13-diHOME as a new therapeutic target for metabolic diseases. Life Sci 2021; 290:120229. [PMID: 34914931 DOI: 10.1016/j.lfs.2021.120229] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 01/23/2023]
Abstract
Lipokines are bioactive compounds, derived from adipose tissue depots, that control several molecular signaling pathways. Recently, 12,13-dihydroxy-9Z-octadecenoic acid (12,13-diHOME), an oxylipin, has gained prominence in the scientific literature. An increase in circulating 12,13-diHOME has been associated with improved metabolic health, and the action of this molecule appears to be mediated by brown adipose tissue (BAT). Scientific evidence indicates that the increase in serum levels of 12,13-diHOME caused by stimuli such as physical exercise and exposure to cold may favor the absorption of fatty acids by brown adipose tissue and stimulate the browning process in white adipose tissue (WAT). Thus, strategies capable of increasing 12,13-diHOME levels may be promising for the prevention and treatment of obesity and metabolic diseases. This review explores the relationship of 12,13-diHOME with brown adipose tissue and its role in the metabolic health context, as well as the signaling pathways involved between 12,13-diHOME and BAT.
Collapse
Affiliation(s)
- Ana Paula Azevêdo Macêdo
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Vitor Rosetto Muñoz
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Dennys Esper Cintra
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, São Paulo, Brazil; Laboratory of Nutritional Genomics, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - José Rodrigo Pauli
- Laboratory of Molecular Biology of Exercise, University of Campinas (UNICAMP), Limeira, São Paulo, Brazil; Laboratory of Cell Signaling, Obesity and Comorbidities Research Center (OCRC), University of Campinas, Campinas, São Paulo, Brazil.
| |
Collapse
|
30
|
Interactions between insulin and exercise. Biochem J 2021; 478:3827-3846. [PMID: 34751700 DOI: 10.1042/bcj20210185] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 02/06/2023]
Abstract
The interaction between insulin and exercise is an example of balancing and modifying the effects of two opposing metabolic regulatory forces under varying conditions. While insulin is secreted after food intake and is the primary hormone increasing glucose storage as glycogen and fatty acid storage as triglycerides, exercise is a condition where fuel stores need to be mobilized and oxidized. Thus, during physical activity the fuel storage effects of insulin need to be suppressed. This is done primarily by inhibiting insulin secretion during exercise as well as activating local and systemic fuel mobilizing processes. In contrast, following exercise there is a need for refilling the fuel depots mobilized during exercise, particularly the glycogen stores in muscle. This process is facilitated by an increase in insulin sensitivity of the muscles previously engaged in physical activity which directs glucose to glycogen resynthesis. In physically trained individuals, insulin sensitivity is also higher than in untrained individuals due to adaptations in the vasculature, skeletal muscle and adipose tissue. In this paper, we review the interactions between insulin and exercise during and after exercise, as well as the effects of regular exercise training on insulin action.
Collapse
|
31
|
Mitochondrial dysfunction plays an essential role in remodeling aging adipose tissue. Mech Ageing Dev 2021; 200:111598. [PMID: 34762939 DOI: 10.1016/j.mad.2021.111598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/12/2021] [Accepted: 11/03/2021] [Indexed: 11/22/2022]
Abstract
Aging is characterized by several physiological changes in the human body, such as the remodeling/redistribution of body fat, highlighted by the increase in fat in the abdominal region due to reduced fat in the peripheral limbs. Abdominal fat is related to metabolic complications and an increased risk for developing diseases such as obesity, type 2 diabetes mellitus, and hypertension. Understanding this process is crucial for developing new therapeutic strategies able to mitigate its impact. This redistribution of fat has been associated with lower activation of brown adipose tissue over the years of life. Brown adipose tissue differs from white adipose tissue, mainly because it produces heat, increasing energy expenditure. Current evidence points to morphological and functional changes in mitochondria during aging, a key mechanism for understanding the dysmetabolic and pro-inflammatory phenotype associated with senescence. Therefore, this minireview will focus on how aging-induced mitochondrial changes are involved in the remodeling/redistribution of body fat.
Collapse
|
32
|
Soppert J, Frisch J, Wirth J, Hemmers C, Boor P, Kramann R, Vondenhoff S, Moellmann J, Lehrke M, Hohl M, van der Vorst EPC, Werner C, Speer T, Maack C, Marx N, Jankowski J, Roma LP, Noels H. A systematic review and meta-analysis of murine models of uremic cardiomyopathy. Kidney Int 2021; 101:256-273. [PMID: 34774555 DOI: 10.1016/j.kint.2021.10.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 09/22/2021] [Accepted: 10/18/2021] [Indexed: 02/06/2023]
Abstract
Chronic kidney disease (CKD) triggers the risk of developing uremic cardiomyopathy as characterized by cardiac hypertrophy, fibrosis and functional impairment. Traditionally, animal studies are used to reveal the underlying pathological mechanism, although variable CKD models, mouse strains and readouts may reveal diverse results. Here, we systematically reviewed 88 studies and performed meta-analyses of 52 to support finding suitable animal models for future experimental studies on pathological kidney-heart crosstalk during uremic cardiomyopathy. We compared different mouse strains and the direct effect of CKD on cardiac hypertrophy, fibrosis and cardiac function in "single hit" strategies as well as cardiac effects of kidney injury combined with additional cardiovascular risk factors in "multifactorial hit" strategies. In C57BL/6 mice, CKD was associated with a mild increase in cardiac hypertrophy and fibrosis and marginal systolic dysfunction. Studies revealed high variability in results, especially regarding hypertrophy and systolic function. Cardiac hypertrophy in CKD was more consistently observed in 129/Sv mice, which express two instead of one renin gene and more consistently develop increased blood pressure upon CKD induction. Overall, "multifactorial hit" models more consistently induced cardiac hypertrophy and fibrosis compared to "single hit" kidney injury models. Thus, genetic factors and additional cardiovascular risk factors can "prime" for susceptibility to organ damage, with increased blood pressure, cardiac hypertrophy and early cardiac fibrosis more consistently observed in 129/Sv compared to C57BL/6 strains.
Collapse
Affiliation(s)
- Josefin Soppert
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany
| | - Janina Frisch
- Department of Biophysics, Center for Human and Molecular Biology (ZHMB), Saarland University, Homburg, Germany
| | - Julia Wirth
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany
| | - Christian Hemmers
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany
| | - Peter Boor
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany; Department of Nephrology and Clinical Immunology, University Hospital RWTH Aachen, Aachen, Germany
| | - Rafael Kramann
- Department of Nephrology and Clinical Immunology, University Hospital RWTH Aachen, Aachen, Germany
| | - Sonja Vondenhoff
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany
| | - Julia Moellmann
- Department of Internal Medicine I, Cardiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Michael Lehrke
- Department of Internal Medicine I, Cardiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Mathias Hohl
- Department of Internal Medicine III, Cardiology/Angiology, University of Homburg, Homburg/Saar, Germany
| | - Emiel P C van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany; Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands; Interdisciplinary Centre for Clinical Research (IZKF), RWTH Aachen University, Aachen, Germany; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany; German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Christian Werner
- Department of Internal Medicine III, Cardiology/Angiology, University of Homburg, Homburg/Saar, Germany
| | - Thimoteus Speer
- Translational Cardio-Renal Medicine, Saarland University, Homburg/Saar, Germany
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Hospital Würzburg, Würzburg, Germany
| | - Nikolaus Marx
- Department of Internal Medicine I, Cardiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Joachim Jankowski
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany; Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Leticia Prates Roma
- Department of Biophysics, Center for Human and Molecular Biology (ZHMB), Saarland University, Homburg, Germany
| | - Heidi Noels
- Institute for Molecular Cardiovascular Research (IMCAR), University Hospital RWTH Aachen, Aachen, Germany; Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
33
|
Cavalcanti-de-Albuquerque JP, Donato J. Rolling out physical exercise and energy homeostasis: Focus on hypothalamic circuitries. Front Neuroendocrinol 2021; 63:100944. [PMID: 34425188 DOI: 10.1016/j.yfrne.2021.100944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/11/2021] [Accepted: 08/18/2021] [Indexed: 01/17/2023]
Abstract
Energy balance is the fine regulation of energy expenditure and energy intake. Negative energy balance causes body weight loss, while positive energy balance promotes weight gain. Modern societies offer a maladapted way of life, where easy access to palatable foods and the lack of opportunities to perform physical activity are considered the roots of the obesity pandemic. Physical exercise increases energy expenditure and, consequently, is supposed to promote weight loss. Paradoxically, physical exercise acutely drives anorexigenic-like effects, but the mechanisms are still poorly understood. Using an evolutionary background, this review aims to highlight the potential involvement of the melanocortin system and other hypothalamic neural circuitries regulating energy balance during and after physical exercise. The physiological significance of these changes will be explored, and possible signalling agents will be addressed. The knowledge discussed here might be important for clarifying obesity aetiology as well as new therapeutic approaches for body weight loss.
Collapse
Affiliation(s)
| | - José Donato
- Department of Physiology and Biophysics, University of São Paulo, São Paulo 05508-900, Brazil.
| |
Collapse
|
34
|
Maharjan BR, Martinez‐Huenchullan SF, Mclennan SV, Twigg SM, Williams PF. Exercise induces favorable metabolic changes in white adipose tissue preventing high-fat diet obesity. Physiol Rep 2021; 9:e14929. [PMID: 34405572 PMCID: PMC8371352 DOI: 10.14814/phy2.14929] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 05/19/2021] [Indexed: 01/08/2023] Open
Abstract
Diet and/or exercise are cost effective interventions to treat obesity. However, it is unclear if the type of exercise undertaken can prevent the onset of obesity and if it can act through different effects on fat depots. In this study we did not allow obesity to develop so we commenced the high-fat diet (HFD) and exercise programs concurrently and investigated the effect of endurance exercise (END) and high-intensity interval training (HIIT) on changes in cellular adipogenesis, thermogenesis, fibrosis, and inflammatory markers in three different fat depots, on a HFD and a chow diet. This was to assess the effectiveness of exercise to prevent the onset of obesity-induced changes. Mice fed with chow or HFD (45% kcal fat) were trained and performed either END or HIIT for 10 weeks (3 x 40 min sessions/week). In HFD mice, both exercise programs significantly prevented the increase in body weight (END: 17%, HIIT: 20%), total body fat mass (END: 46%, HIIT: 50%), increased lean mass as a proportion of body weight (Lean mass/BW) by 14%, and improved insulin sensitivity by 22%. Further evidence of the preventative effect of exercise was seen significantly decreased markers for adipogenesis, inflammation, and extracellular matrix accumulation in both subcutaneous adipose tissue (SAT) and epididymal adipose tissue (EPI). In chow, no such marked effects were seen with both the exercise programs on all the three fat depots. This study establishes the beneficial effect of both HIIT and END exercise in preventing metabolic deterioration, collagen deposition, and inflammatory responses in fat depots, resulting in an improved whole body insulin resistance in HFD mice.
Collapse
Affiliation(s)
- Babu R. Maharjan
- Greg Brown Diabetes & Endocrinology LaboratorySydney Medical SchoolUniversity of SydneySydneyAustralia
- Department of BiochemistryPatan Academy of Health SciencesSchool of MedicineLalitpurNepal
| | - Sergio F. Martinez‐Huenchullan
- Greg Brown Diabetes & Endocrinology LaboratorySydney Medical SchoolUniversity of SydneySydneyAustralia
- Faculty of MedicineSchool of Physical TherapyUniversidad Austral de ChileValdiviaChile
| | - Susan V. Mclennan
- Greg Brown Diabetes & Endocrinology LaboratorySydney Medical SchoolUniversity of SydneySydneyAustralia
- New South Wales Health PathologySydneyAustralia
- Department of EndocrinologyRoyal Prince Alfred HospitalSydneyAustralia
| | - Stephen M. Twigg
- Greg Brown Diabetes & Endocrinology LaboratorySydney Medical SchoolUniversity of SydneySydneyAustralia
- Department of EndocrinologyRoyal Prince Alfred HospitalSydneyAustralia
| | - Paul F. Williams
- Greg Brown Diabetes & Endocrinology LaboratorySydney Medical SchoolUniversity of SydneySydneyAustralia
- New South Wales Health PathologySydneyAustralia
- Department of EndocrinologyRoyal Prince Alfred HospitalSydneyAustralia
| |
Collapse
|
35
|
McKie GL, Shamshoum H, Hunt KL, Thorpe HHA, Dibe HA, Khokhar JY, Doucette CA, Wright DC. Intermittent cold exposure improves glucose homeostasis despite exacerbating diet-induced obesity in mice housed at thermoneutrality. J Physiol 2021; 600:829-845. [PMID: 34192813 DOI: 10.1113/jp281774] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/28/2021] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Ambient cold exposure is often regarded as a promising anti-obesity treatment in mice. However, most preclinical studies aimed at treating obesity via cold-induced thermogenesis have been confounded by subthermoneutral housing temperatures. Therefore, the ability of ambient cold to combat diet-induced obesity in mice housed under humanized thermoneutral conditions is currently unknown. Moreover, mammals such as mice are rarely exposed to chronic ambient cold without reprieve, yet mice are often subjected to experimental conditions of chronic rather than intermittent cold exposure (ICE), despite ICE being more physiologically relevant. In the present study, we provide novel evidence that thermoneutral housing uncouples the effects of ICE on glucose and energy homeostasis suggesting that ICE, despite improving glucose tolerance, is not an effective obesity treatment when mice are housed under humanized thermoneutral conditions. ABSTRACT The present study examines whether a physiologically relevant model of ambient cold exposure, intermittent cold exposure (ICE), could ameliorate the metabolic impairments of diet-induced obesity in male and female mice housed under humanized thermoneutral conditions. Male and female C57BL/6J mice housed at thermoneutrality (29°C) were fed a low-fat diet or high-fat diet for 6 weeks before being weight matched into groups that remained unperturbed or underwent ICE for 4 weeks (4°C for 60 min day-1 ; 5 days week-1 ) when being maintained on their respective diets. ICE induced rapid and persistent hyperphagia exacerbating rather than attenuating high-fat diet-induced obesity over time. These ICE-induced increases in adiposity were found to be energy intake-dependent via pair-feeding. Despite exacerbating high-fat diet-induced obesity, ICE improved glucose tolerance, independent of diet, in a sex-specific manner. The effects of ICE on glucose tolerance were not attributed to improvements in whole-body insulin tolerance, tissue specific insulin action, nor differences in markers of hepatic insulin clearance or pancreatic beta cell proliferation. Instead, ICE increased serum concentrations of insulin and C-peptide in response to glucose, suggesting that ICE may improve glucose tolerance by potentiating pancreatic glucose-stimulated insulin secretion. These data suggest that ICE, despite improving glucose tolerance, is not an effective obesity treatment in mice housed under humanized conditions.
Collapse
Affiliation(s)
- Greg L McKie
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Hesham Shamshoum
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Kristin L Hunt
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
| | - Hayley H A Thorpe
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
| | - Hana A Dibe
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| | - Jibran Y Khokhar
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
| | - Christine A Doucette
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, MB, Canada
| | - David C Wright
- Department of Human Health & Nutritional Sciences, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
36
|
Gene Expression Analysis of Environmental Temperature and High-Fat Diet-Induced Changes in Mouse Supraclavicular Brown Adipose Tissue. Cells 2021; 10:cells10061370. [PMID: 34199472 PMCID: PMC8226907 DOI: 10.3390/cells10061370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 05/26/2021] [Accepted: 05/29/2021] [Indexed: 12/16/2022] Open
Abstract
Obesity, a dysregulation of adipose tissue, is a major health risk factor associated with many diseases. Brown adipose tissue (BAT)-mediated thermogenesis can potentially regulate energy expenditure, making it an attractive therapeutic target to combat obesity. Here, we characterize the effects of cold exposure, thermoneutrality, and high-fat diet (HFD) feeding on mouse supraclavicular BAT (scBAT) morphology and BAT-associated gene expression compared to other adipose depots, including the interscapular BAT (iBAT). scBAT was as sensitive to cold induced thermogenesis as iBAT and showed reduced thermogenic effect under thermoneutrality. While both scBAT and iBAT are sensitive to cold, the expression of genes involved in nutrient processing is different. The scBAT also showed less depot weight gain and more single-lipid adipocytes, while the expression of BAT thermogenic genes, such as Ucp1, remained similar or increased more under our HFD feeding regime at ambient and thermoneutral temperatures than iBAT. Together, these findings show that, in addition to its anatomical resemblance to human scBAT, mouse scBAT possesses thermogenic features distinct from those of other adipose depots. Lastly, this study also characterizes a previously unknown mouse deep neck BAT (dnBAT) depot that exhibits similar thermogenic characteristics as scBAT under cold exposure and thermoneutrality.
Collapse
|
37
|
Normann MC, Cox M, Akinbo OI, Watanasriyakul WT, Kovalev D, Ciosek S, Miller T, Grippo AJ. Differential paraventricular nucleus activation and behavioral responses to social isolation in prairie voles following environmental enrichment with and without physical exercise. Soc Neurosci 2021; 16:375-390. [PMID: 33947321 DOI: 10.1080/17470919.2021.1926320] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Social stressors produce neurobiological and emotional consequences in social species. Environmental interventions, such as environmental enrichment and exercise, may modulate physiological and behavioral stress responses. The present study investigated the benefits of environmental enrichment and exercise against social stress in the socially monogamous prairie vole. Female prairie voles remained paired with a sibling (control) or were isolated from a sibling for 4 weeks. The isolated groups were assigned to isolated sedentary, isolated with environmental enrichment, or isolated with both enrichment and exercise conditions. Behaviors related to depression, anxiety, and sociality were investigated using the forced swim test (FST), elevated plus maze (EPM), and a social crowding stressor (SCS), respectively. cFos expression was evaluated in stress-related circuitry following the SCS. Both enrichment and enrichment with exercise protected against depression-relevant behaviors in the FST and social behavioral disruptions in the SCS, but only enrichment with exercise protected against anxiety-related behaviors in the EPM and altered cFos expression in the hypothalamic paraventricular nucleus in isolated prairie voles. Enrichment may improve emotion-related and social behaviors, however physical exercise may be an important component of environmental strategies for protecting against anxiety-related behaviors and reducing neural activation as a function of social stress.
Collapse
Affiliation(s)
- Marigny C Normann
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | - Miranda Cox
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | - Oreoluwa I Akinbo
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | | | - Dmitry Kovalev
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | - Sarah Ciosek
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | - Thomas Miller
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| | - Angela J Grippo
- Department of Psychology, Northern Illinois University, DeKalb, IL, USA
| |
Collapse
|
38
|
Clart LM, Welly RJ, Queathem ED, Rector RS, Padilla J, Baines CP, Kanaley JA, Lubahn DB, Vieira-Potter VJ. Role of ERβ in adipocyte metabolic response to wheel running following ovariectomy. J Endocrinol 2021; 249:223-237. [PMID: 33877054 PMCID: PMC8713017 DOI: 10.1530/joe-21-0009] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/20/2021] [Indexed: 11/08/2022]
Abstract
Estrogen receptor β (ERb), one of the two major estrogen receptors, acts via genomic and non-genomic signaling pathways to affect many metabolic functions, including mitochondrial biogenesis and respiration. This study assessed the effect of ERb classical genomic activity on adipocyte-specific and -systemic metabolic responses to wheel running exercise in a rodent model of menopause. Female mice lacking the ERb DNA-binding domain (ERbDBDKO, n = 20) and WT (n = 21) littermate controls were fed a high-fat diet (HFD), ovariectomized (OVX), and randomized to control (no running wheel) and exercise (running wheel access) groups and were followed for 8 weeks. Wheel running did not confer protection against metabolic dysfunction associated with HFD+OVX in either ERbDBDKO or WT mice, despite increased energy expenditure. Unexpectedly, in the ERbDBDKO group, wheel running increased fasting insulin and surrogate measures of insulin resistance, and modestly increased adipose tissue inflammatory gene expression (P ≤ 0.05). These changes were not accompanied by significant changes in adipocyte mitochondrial respiration. It was demonstrated for the first time that female WT OVX mice do experience exercise-induced browning of white adipose tissue, indicated by a robust increase in uncoupling protein 1 (UCP1) (P ≤ 0.05). However, KO mice were completely resistant to this effect, indicating that full ERb genomic activity is required for exercise-induced browning. The inability to upregulate UCP1 with exercise following OVX may have resulted in the increased insulin resistance observed in KO mice, a hypothesis requiring further investigation.
Collapse
Affiliation(s)
- Laura M Clart
- Department of Nutrition and Exercise Physiology, University of Missouri System, Columbia, Missouri, USA
| | - Rebecca J Welly
- Department of Nutrition and Exercise Physiology, University of Missouri System, Columbia, Missouri, USA
| | - Eric D Queathem
- Department of Nutrition and Exercise Physiology, University of Missouri System, Columbia, Missouri, USA
| | - R Scott Rector
- Department of Nutrition and Exercise Physiology, University of Missouri System, Columbia, Missouri, USA
- Internal Medicine-Division of Gastroenterology and Hepatology, University of Missouri System, Columbia, Missouri, USA
- Research Service, Truman VA Memorial Hospital, Columbia, Missouri, USA
| | - Jaume Padilla
- Department of Nutrition and Exercise Physiology, University of Missouri System, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri System System, Columbia, Missouri, USA
| | - Christopher P Baines
- Department of Biomedical Sciences, University of Missouri System, Columbia, Missouri, USA
| | - Jill A Kanaley
- Department of Nutrition and Exercise Physiology, University of Missouri System, Columbia, Missouri, USA
| | - Dennis B Lubahn
- Department of Biochemistry, University of Missouri System, Columbia, Missouri, USA
| | - Victoria J Vieira-Potter
- Department of Nutrition and Exercise Physiology, University of Missouri System, Columbia, Missouri, USA
| |
Collapse
|
39
|
Lu S, Wei F, Li G. The evolution of the concept of stress and the framework of the stress system. Cell Stress 2021; 5:76-85. [PMID: 34124582 PMCID: PMC8166217 DOI: 10.15698/cst2021.06.250] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 12/13/2022] Open
Abstract
Stress is a central concept in biology and has now been widely used in psychological, physiological, social, and even environmental fields. However, the concept of stress was cross-utilized to refer to different elements of the stress system including stressful stimulus, stressor, stress response, and stress effect. Here, we summarized the evolution of the concept of stress and the framework of the stress system. We find although the concept of stress is developed from Selye's "general adaptation syndrome", it has now expanded and evolved significantly. Stress is now defined as a state of homeostasis being challenged, including both system stress and local stress. A specific stressor may potentially bring about specific local stress, while the intensity of stress beyond a threshold may commonly activate the hypothalamic-pituitary-adrenal axis and result in a systematic stress response. The framework of the stress system indicates that stress includes three types: sustress (inadequate stress), eustress (good stress), and distress (bad stress). Both sustress and distress might impair normal physiological functions and even lead to pathological conditions, while eustress might benefit health through hormesis-induced optimization of homeostasis. Therefore, an optimal stress level is essential for building biological shields to guarantee normal life processes.
Collapse
Affiliation(s)
- Siyu Lu
- Center for Aging Biomedicine, Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Fang Wei
- Center for Aging Biomedicine, Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
- National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Guolin Li
- Center for Aging Biomedicine, Key Laboratory of Protein Chemistry and Developmental Biology of Fish of Ministry of Education, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
- National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
- Key Laboratory of Hunan Province for Model Animal and Stem Cell Biology, School of Medicine, Hunan Normal University, Changsha, Hunan 410081, China
| |
Collapse
|
40
|
Li L, Huang C, Yin H, Zhang X, Wang D, Ma C, Li J, Zhao Y, Li X. Interleukin-6 mediated exercise-induced alleviation of adiposity and hepatic steatosis in mice. BMJ Open Diabetes Res Care 2021; 9:9/1/e001431. [PMID: 33853848 PMCID: PMC8054088 DOI: 10.1136/bmjdrc-2020-001431] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 02/18/2021] [Accepted: 03/21/2021] [Indexed: 12/25/2022] Open
Abstract
INTRODUCTION Exercise training has been shown to be the most effective strategy to combat obesity and non-alcoholic fatty liver disease. However, exercise promotes loss of adipose tissue mass and improves obesity-related hepatic steatosis through mechanisms that remain obscure. RESEARCH DESIGN AND METHODS To study the role of interleukin-6 (IL-6) in high-fat diet (HFD)-induced adiposity and hepatic steatosis during treadmill running, IL-6 knockout (IL-6 KO) mice and wild-type (WT) mice were randomly divided into lean, obese (fed a HFD) and trained obese groups (fed a HFD and exercise trained). RESULTS After 20 weeks of HFD feeding and 8 weeks of treadmill running, we found that exercise obviously reduced HFD-induced body weight gain, inhibited visceral adipose tissue (VAT) and subcutaneous adipose tissue (SAT) expansion and almost completely reversed obesity-related intrahepatic fat accumulation in WT mice. However, IL-6 knockout (IL-6 KO) mice are refractory to the benefits of treadmill training on body weight, VAT and SAT mass elevation, and hepatic steatosis. Moreover, a panel of lipolytic-related and thermogenic-related genes, including ATGL, HSL and PGC-1α, was upregulated in the VAT and SAT of WT mice that received exercise training compared with untrained mice, which was not observed in IL-6 KO mice. In addition, exercise training resulted in a significant inhibition of hepatic peroxisome proliferator-activated receptor gamma (PPAR-γ) expression in WT mice, and these effects were not noted in IL-6 KO mice. CONCLUSION These results revealed that IL-6 is involved in the prevention of obesity and hepatic fat accumulation during exercise training. The mechanisms underlying these antiobesity effects may be associated with enhanced lipolysis and thermogenesis in white adipose tissue. The improvement in hepatic steatosis by exercise training may benefit from the marked inhibition of PPAR-γ expression by IL-6.
Collapse
Affiliation(s)
- Long Li
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
- Institute of Drug Discovery Technology, Ningbo University, Ningbo, China
| | - Caoxin Huang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Hongyan Yin
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Xiaofang Zhang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Dongmei Wang
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Chen Ma
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Jia Li
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Yan Zhao
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| | - Xuejun Li
- Department of Endocrinology and Diabetes, Xiamen Diabetes Institute, Fujian Key Laboratory of Translational Research for Diabetes, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
41
|
Allerton TD, Kowalski GM, Stampley J, Irving BA, Lighton JRB, Floyd ZE, Stephens JM. An Ethanolic Extract of Artemisia dracunculus L. Enhances the Metabolic Benefits of Exercise in Diet-induced Obese Mice. Med Sci Sports Exerc 2021; 53:712-723. [PMID: 33105388 PMCID: PMC9045727 DOI: 10.1249/mss.0000000000002516] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE The purpose of this study was to determine the effect of an ethanolic extract of Artemisia dracunculus L. (5011) combined with exercise on in vivo glucose and fat metabolism in diet-induced obese male mice. METHODS After 8 wk of high-fat diet (HFD) feeding, 52 mice were randomly allocated to a voluntary wheel running group (HFD Ex), a 5011 + HFD sedentary group (5011 Sed), a 5011 + HFD Ex (5011 Ex), or an HFD sedentary group (HFD Sed) for 4 wk. Real-time energy expenditure and substrate utilization were measured by indirect calorimetry. A stable isotope glucose tolerance test was performed before and after the 4-wk wheel running period to determine changes in endogenous glucose production and glucose disposal. We also performed an analysis of genes and proteins associated with the early response to exercise and exercise adaptations in skeletal muscle and liver. RESULTS When compared with HFD Ex mice, 5011 Ex mice had increased fat oxidation during speed- and distance-matched wheel running bouts. Both HFD Ex and 5011 Ex mice had reduced endogenous glucose during the glucose tolerance test, whereas only the 5011 Sed and the 5011 Ex mice had improved glucose disposal after the 4-wk experimental period when compared with HFD Sed and HFD Ex mice. 5011 Ex mice had increased Pgc1-α and Tfam expression in skeletal muscle when compared with HFD Ex mice, whereas Pdk4 expression was reduced in the liver of HFD Ex and 5011 Ex mice. CONCLUSIONS Our study demonstrates that 5011, an ethanolic extract of A. dracunculus L., with a history of medicinal use, enhances the metabolic benefits of exercise to improve in vivo fat and glucose metabolism.
Collapse
Affiliation(s)
| | - Greg M Kowalski
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, AUSTRALIA
| | - James Stampley
- Department of Kinesiology, Louisiana State University, Baton Rouge, LA
| | - Brian A Irving
- Department of Kinesiology, Louisiana State University, Baton Rouge, LA
| | | | | | | |
Collapse
|
42
|
Exercise-A Panacea of Metabolic Dysregulation in Cancer: Physiological and Molecular Insights. Int J Mol Sci 2021; 22:ijms22073469. [PMID: 33801684 PMCID: PMC8037630 DOI: 10.3390/ijms22073469] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 03/21/2021] [Accepted: 03/24/2021] [Indexed: 12/14/2022] Open
Abstract
Metabolic dysfunction is a comorbidity of many types of cancers. Disruption of glucose metabolism is of concern, as it is associated with higher cancer recurrence rates and reduced survival. Current evidence suggests many health benefits from exercise during and after cancer treatment, yet only a limited number of studies have addressed the effect of exercise on cancer-associated disruption of metabolism. In this review, we draw on studies in cells, rodents, and humans to describe the metabolic dysfunctions observed in cancer and the tissues involved. We discuss how the known effects of acute exercise and exercise training observed in healthy subjects could have a positive outcome on mechanisms in people with cancer, namely: insulin resistance, hyperlipidemia, mitochondrial dysfunction, inflammation, and cachexia. Finally, we compile the current limited knowledge of how exercise corrects metabolic control in cancer and identify unanswered questions for future research.
Collapse
|
43
|
Fuller KNZ, Thyfault JP. Barriers in translating preclinical rodent exercise metabolism findings to human health. J Appl Physiol (1985) 2021; 130:182-192. [PMID: 33180643 PMCID: PMC7944931 DOI: 10.1152/japplphysiol.00683.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 10/21/2020] [Accepted: 11/10/2020] [Indexed: 01/03/2023] Open
Abstract
Physical inactivity and low aerobic capacity are primary drivers of chronic disease pathophysiology and are independently associated with all-cause mortality. Conversely, increased physical activity and exercise are central to metabolic disease prevention and longevity. Although these relationships are well characterized in the literature, what remains incompletely understood are the mechanisms by which physical activity/exercise prevents disease. Given methodological constraints of clinical research, investigators must often rely on preclinical rodent models to investigate these potential underlying mechanisms. However, there are several key barriers to applying exercise metabolism findings from rodent models to human health. These barriers include housing temperature, nutrient metabolism, exercise modality, exercise testing, and sex differences. Increased awareness and understanding of these barriers will enhance the ability to impact human health through more appropriate experimental design and interpretation of data within the context of these factors.
Collapse
Affiliation(s)
- Kelly N Z Fuller
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
| | - John P Thyfault
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, Kansas
- Division of Endocrinology and Metabolism, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas
- Research Service Kansas City Veterans Affairs Medical Center, Kansas City, Kansas
- Center for Children's Healthy Lifestyles and Nutrition, Kansas City, Missouri
| |
Collapse
|
44
|
Munten S, Ménard L, Gagnon J, Dorman SC, Mezouari A, Gagnon DD. High-intensity interval exercise in the cold regulates acute and postprandial metabolism. J Appl Physiol (1985) 2020; 130:408-420. [PMID: 33270513 DOI: 10.1152/japplphysiol.00384.2020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
High-intensity interval exercise (HIIE) has been shown to be more effective than moderate-intensity exercise for increasing acute lipid oxidation and lowering blood lipids during exercise and postprandially. Exercise in cold environments is also known to enhance lipid oxidation; however, the immediate and long-term effects of HIIE exercise in cold are unknown. The purpose of this study was to examine the effects cold stress during HIIE on acute exercise metabolism and postprandial metabolism. Eleven recreationally active individuals (age: 23 ± 3 yr, weight: 80 ± 9.7 kg, V̇O2peak: 39.2 ± 5.73 mL·kg-1·min-1) performed evening HIIE sessions (10 × 60 s cycling, 90% V̇O2peak interspersed with 90 s active recovery, 30% V̇O2peak) in thermoneutral (HIIE-TN, control; 21°C) and cold environment (HIIE-CO; 0°C), following a balanced crossover design. The following morning, participants consumed a high-fat meal. Indirect calorimetry was used to assess substrate oxidation, and venous blood samples were obtained to assess changes in noncellular metabolites. During acute exercise, lipid oxidation was higher in HIIE-CO (P = 0.002) without differences in V̇O2 and energy expenditure (P ≥ 0.162) between conditions. Postprandial V̇O2, lipid and CHO oxidation, plasma insulin, and triglyceride concentrations were not different between conditions (P > 0.05). Postprandial blood LDL-C levels were higher in HIIE-CO 2 h after the meal (P = 0.003). Postprandial glucose area under curve was 49% higher in HIIE-CO versus HIIE-TN (P = 0.034). Under matched energy expenditure conditions, HIIE demonstrated higher lipid oxidation rates during exercise in the cold; but only marginally influenced postprandial lipid metabolism the following morning. In conclusion, HIIE in the cold seemed to be less favorable for postprandial lipid and glycemic responses.NEW & NOTEWORTHY This is the first known study to investigate the effects of cold ambient temperatures on acute metabolism during high-intensity interval exercise, as well as postprandial metabolism the next day. We observed that high-intensity interval exercise in a cold environment does change acute metabolism compared to a thermoneutral environment; however, the addition of a cold stimulus was less favorable for postprandial metabolic responses the following day.
Collapse
Affiliation(s)
- Stephanie Munten
- Laboratory of Environmental Exercise Physiology, School of Kinesiology and Health Sciences, Laurentian University, Sudbury, Canada.,Centre for Research in Occupational Safety and Health, Laurentian University, Sudbury, Canada
| | - Lucie Ménard
- Laboratory of Environmental Exercise Physiology, School of Kinesiology and Health Sciences, Laurentian University, Sudbury, Canada.,Centre for Research in Occupational Safety and Health, Laurentian University, Sudbury, Canada.,Northern Ontario School of Medicine, Laurentian University, Sudbury, Canada
| | - Jeffrey Gagnon
- Department of Biology, Laurentian University, Sudbury, Canada
| | - Sandra C Dorman
- Laboratory of Environmental Exercise Physiology, School of Kinesiology and Health Sciences, Laurentian University, Sudbury, Canada.,Centre for Research in Occupational Safety and Health, Laurentian University, Sudbury, Canada.,Northern Ontario School of Medicine, Laurentian University, Sudbury, Canada
| | - Ania Mezouari
- Department of Biology, Laurentian University, Sudbury, Canada
| | - Dominique D Gagnon
- Laboratory of Environmental Exercise Physiology, School of Kinesiology and Health Sciences, Laurentian University, Sudbury, Canada.,Centre for Research in Occupational Safety and Health, Laurentian University, Sudbury, Canada
| |
Collapse
|
45
|
Amano Y, Nonaka Y, Takeda R, Kano Y, Hoshino D. Effects of electrical stimulation-induced resistance exercise training on white and brown adipose tissues and plasma meteorin-like concentration in rats. Physiol Rep 2020; 8:e14540. [PMID: 32812347 PMCID: PMC7435032 DOI: 10.14814/phy2.14540] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/16/2020] [Accepted: 07/20/2020] [Indexed: 12/26/2022] Open
Abstract
Chronic endurance exercise training induces morphological and metabolic alterations including mitochondrial biogenesis in white adipose tissue (WAT) and brown adipose tissue (BAT) in rodents. A myokine called meteorin-like (Metrnl) is associated with morphological and metabolic adaptation and increased in blood after acute resistance exercise. However, the effects of chronic resistance exercise training (RT), which aims to increase muscle mass and strength, on WAT and BAT are unclear. Therefore, we aimed to clarify the effects of RT on morphological and metabolic parameters in WAT and BAT and on plasma Metrnl concentrations. We applied electrical stimulation to both legs of rats as RT three times a week for 4 weeks. RT reduced adipocyte size in subcutaneous WAT but induced no changes in mitochondrial and thermogenesis proteins. In BAT, peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) protein levels and mitochondrial content markers were significantly higher in the RT group compared with the control group. A significant positive correlation was found between the expression of PGC-1α in BAT and plasma Metrnl concentrations. These results suggest that plasma Metrnl is associated with PGC-1α and mitochondrial biogenesis in BAT. This study describes a potential role of RT in preventing metabolic diseases via altering WAT and BAT and increasing plasma Mertnl concentrations.
Collapse
Affiliation(s)
- Yuhei Amano
- Bioscience and Technology ProgramDepartment of Engineering ScienceThe University of Electro‐CommunicationsChofuJapan
| | - Yudai Nonaka
- Bioscience and Technology ProgramDepartment of Engineering ScienceThe University of Electro‐CommunicationsChofuJapan
- Japan Society for the Promotion of Science (JSPS)TokyoJapan
| | - Reo Takeda
- Bioscience and Technology ProgramDepartment of Engineering ScienceThe University of Electro‐CommunicationsChofuJapan
| | - Yutaka Kano
- Bioscience and Technology ProgramDepartment of Engineering ScienceThe University of Electro‐CommunicationsChofuJapan
| | - Daisuke Hoshino
- Bioscience and Technology ProgramDepartment of Engineering ScienceThe University of Electro‐CommunicationsChofuJapan
| |
Collapse
|
46
|
Inducible deletion of skeletal muscle AMPKα reveals that AMPK is required for nucleotide balance but dispensable for muscle glucose uptake and fat oxidation during exercise. Mol Metab 2020; 40:101028. [PMID: 32504885 PMCID: PMC7356270 DOI: 10.1016/j.molmet.2020.101028] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 02/05/2023] Open
Abstract
Objective Evidence for AMP-activated protein kinase (AMPK)-mediated regulation of skeletal muscle metabolism during exercise is mainly based on transgenic mouse models with chronic (lifelong) disruption of AMPK function. Findings based on such models are potentially biased by secondary effects related to a chronic lack of AMPK function. To study the direct effect(s) of AMPK on muscle metabolism during exercise, we generated a new mouse model with inducible muscle-specific deletion of AMPKα catalytic subunits in adult mice. Methods Tamoxifen-inducible and muscle-specific AMPKα1/α2 double KO mice (AMPKα imdKO) were generated by using the Cre/loxP system, with the Cre under the control of the human skeletal muscle actin (HSA) promoter. Results During treadmill running at the same relative exercise intensity, AMPKα imdKO mice showed greater depletion of muscle ATP, which was associated with accumulation of the deamination product IMP. Muscle-specific deletion of AMPKα in adult mice promptly reduced maximal running speed and muscle glycogen content and was associated with reduced expression of UGP2, a key component of the glycogen synthesis pathway. Muscle mitochondrial respiration, whole-body substrate utilization, and muscle glucose uptake and fatty acid (FA) oxidation during muscle contractile activity remained unaffected by muscle-specific deletion of AMPKα subunits in adult mice. Conclusions Inducible deletion of AMPKα subunits in adult mice reveals that AMPK is required for maintaining muscle ATP levels and nucleotide balance during exercise but is dispensable for regulating muscle glucose uptake, FA oxidation, and substrate utilization during exercise. Inducible deletion of AMPKα in adult mice disturbs nucleotide balance during exercise. Inducible deletion of AMPKα in adult mice lowers muscle glycogen content and reduces exercise capacity. Muscle mitochondrial respiration, and glucose uptake and FA oxidation during muscle contractions remain unaffected by AMPKα deletion.
Collapse
|
47
|
Severinsen MCK, Schéele C, Pedersen BK. Exercise and browning of white adipose tissue - a translational perspective. Curr Opin Pharmacol 2020; 52:18-24. [PMID: 32480032 DOI: 10.1016/j.coph.2020.04.004] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/16/2020] [Accepted: 04/19/2020] [Indexed: 12/26/2022]
Abstract
Browning of white adipose tissue is a cold-induced phenomenon in rodents, constituted by the differentiation of a subset of thermogenic adipocytes among existing white adipocytes. Emerging evidence in the literature points at additional factors and environmental conditions stimulating browning in rodents, including physical exercise training. Exercise engages sympathetic activation which during cold activation promotes proliferation and differentiation of brown preadipocytes. Exercise also stimulates the release of multiple growth factors and cytokines. Importantly, there are clear discrepancies between human and rodents with regard to thermogenic capacity and browning potential. Here we provide a translational perspective on exercise-induced browning and review recent findings on the role of myokines and hepatokines in this process.
Collapse
Affiliation(s)
- Mai Charlotte Krogh Severinsen
- Centre of Inflammation and Metabolism/Centre for Physical Activity Research (CIM/CFAS), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Camilla Schéele
- Centre of Inflammation and Metabolism/Centre for Physical Activity Research (CIM/CFAS), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark; The Novo Nordisk Foundation Center for Basic Metabolic Research, Copenhagen University Hospital, 2200 Copenhagen, Denmark
| | - Bente Klarlund Pedersen
- Centre of Inflammation and Metabolism/Centre for Physical Activity Research (CIM/CFAS), Rigshospitalet, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
48
|
Vidal P, Stanford KI. Exercise-Induced Adaptations to Adipose Tissue Thermogenesis. Front Endocrinol (Lausanne) 2020; 11:270. [PMID: 32411099 PMCID: PMC7201000 DOI: 10.3389/fendo.2020.00270] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 04/14/2020] [Indexed: 12/15/2022] Open
Abstract
Exercise training results in beneficial adaptations to numerous tissues and offers protection against metabolic disorders including obesity and type 2 diabetes. Multiple studies have indicated that both white (WAT) and brown (BAT) adipose tissue may play an important role to mediate the beneficial effects of exercise. Studies from both rodents and humans have identified exercise-induced changes in WAT including increased mitochondrial activity and glucose uptake, an altered endocrine profile, and in rodents, a beiging of the WAT. Studies investigating the effects of exercise on BAT have resulted in conflicting data in terms of mitochondrial activity, glucose uptake, and thermogenic activity in rodents and humans, and remain an important area of investigation. This review discusses the exercise-induced adaptations to white and brown adipose tissue, distinguishing important differences between rodents and humans and highlighting the latest studies in the field and their implications.
Collapse
|
49
|
Halpern B, Mancini MC, Mendes C, Machado CML, Prando S, Sapienza MT, Buchpiguel CA, do Amaral FG, Cipolla-Neto J. Melatonin deficiency decreases brown adipose tissue acute thermogenic capacity of in rats measured by 18F-FDG PET. Diabetol Metab Syndr 2020; 12:82. [PMID: 32973928 PMCID: PMC7504678 DOI: 10.1186/s13098-020-00589-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 09/10/2020] [Indexed: 01/27/2023] Open
Abstract
OBJECTIVE Melatonin has been shown to increase brown adipose tissue (BAT) mass, which can lead to important metabolic effects, such as bodyweight reduction and glycemic improvement. However, BAT mass can only be measured invasively and. The gold standard for non-invasive measurement of BAT activity is positron emission tomography with 2-deoxy-2-[fluorine-18] fluoro-d-glucose (18F-FDG PET). There is no study, to our knowledge, that has evaluated if melatonin influences BAT activity, measured by this imaging technique in animals. METHODS Three experimental groups of Wistar rats (control, pinealectomy, and pinealectomy replaced with melatonin) had an 18F-FDG PET performed at room temperature and after acute cold exposure. The ratio of increased BAT activity after cold exposure/room temperature was called "acute thermogenic capacity" (ATC) We also measured UCP-1 mRNA expression to correlate with the 18F-FDG PET results. RESULTS Pinealectomy led to reduced acute thermogenic capacity, compared with the other groups, as well as reduced UCP1 mRNA expression. CONCLUSION Melatonin deficiency impairs BAT response when exposed to acute cold exposure. These results can lead to future studies of the influence of melatonin on BAT, in animals and humans, without needing an invasive evaluation of BAT.
Collapse
Affiliation(s)
- Bruno Halpern
- Department of Endocrinology and Metabolism, Hospital das Clinicas da Faculdade de Medicina de São Paulo, São Paulo, Brazil
| | - Marcio C. Mancini
- Department of Endocrinology and Metabolism, Hospital das Clinicas da Faculdade de Medicina de São Paulo, São Paulo, Brazil
| | - Caroline Mendes
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, Brazil
| | - Camila Maria Longo Machado
- Nuclear Medicine Institute, Hospital das Clinicas da Faculdade de Medicina de São Paulo, São Paulo, Brazil
| | - Silvana Prando
- Nuclear Medicine Institute, Hospital das Clinicas da Faculdade de Medicina de São Paulo, São Paulo, Brazil
| | - Marcelo Tatit Sapienza
- Nuclear Medicine Institute, Hospital das Clinicas da Faculdade de Medicina de São Paulo, São Paulo, Brazil
| | - Carlos Alberto Buchpiguel
- Nuclear Medicine Institute, Hospital das Clinicas da Faculdade de Medicina de São Paulo, São Paulo, Brazil
| | | | - José Cipolla-Neto
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, Universidade de São Paulo, São Paulo, Brazil
| |
Collapse
|