1
|
Hansen M, Lange KK, Stausholm MB, Dela F. Are Individuals With Type 2 Diabetes Metabolically Inflexible? A Systematic Review and Meta-Analysis. Endocrinol Diabetes Metab 2025; 8:e70044. [PMID: 40318136 PMCID: PMC12048703 DOI: 10.1002/edm2.70044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 02/04/2025] [Accepted: 03/03/2025] [Indexed: 05/07/2025] Open
Abstract
AIM Type 2 diabetes (T2D) is characterised by insulin resistance and possibly by impaired metabolic flexibility, the latter referring to the body's ability to switch between fuel sources. This review systematically examines metabolic flexibility, measured by changes in the respiratory exchange ratio (ΔRER) during hyperinsulinaemic clamps, across lean, overweight/obese, and T2D populations. METHODS A comprehensive search of PubMed identified 65 studies meeting the inclusion criteria, with 35 using a ~40 mU/m2/min insulin infusion rate for accurate comparisons. These studies included 985 participants: 256 lean, 497 overweight/obese, and 232 T2D individuals. The differences in ΔRER between the three groups were meta-analysed. RESULTS Basal RER values did not significantly differ across groups, but insulin-stimulated ΔRER was higher in lean individuals compared to overweight/obese and T2D groups (ΔRER values 0.10, 0.07 and 0.07, respectively; p = 0.037) indicating greater metabolic flexibility in the lean group. However, high statistical heterogeneity in the ΔRER within-group results (I2 values: 92.3%-94.5%) suggests considerable variability among studies. A meta-regression analysis accounting for age, sex, and BMI indicated that only BMI was significantly associated with ΔRER. Factors contributing to the remaining heterogeneity likely include differences in participant characteristics (e.g., glycaemic control) and study design. CONCLUSIONS The review highlights the need for standardised data presentation in metabolic studies. Overall, metabolic flexibility appears more influenced by overweight status than T2D per se, challenging the notion of a distinct metabolic inflexibility threshold for T2D.
Collapse
Affiliation(s)
- Maria Hansen
- Department of Biomedical Sciences, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Kristine Kjær Lange
- Department of Biomedical Sciences, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Martin Bjørn Stausholm
- Department of Physical and Occupational TherapyCopenhagen University Hospital, Bispebjerg and FrederiksbergCopenhagenDenmark
- Department of Global Public Health and Primary CareUniversity of BergenBergenNorway
| | - Flemming Dela
- Department of Biomedical Sciences, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Laboratory of Sports and Nutrition ResearchRiga Stradins UniversityRigaLatvia
| |
Collapse
|
2
|
Ashcroft SP, Ehrlich AM, Burek K, Pendergrast LA, Yonamine CY, Treebak JT, Zierath JR. Enhanced metabolic adaptations following late dark phase wheel running in high-fat diet-fed mice. Mol Metab 2025; 95:102116. [PMID: 39993626 PMCID: PMC11930447 DOI: 10.1016/j.molmet.2025.102116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/19/2025] [Accepted: 02/20/2025] [Indexed: 02/26/2025] Open
Abstract
Exercise interventions represent an effective strategy to prevent and treat metabolic diseases and the time-of-day-dependent effects of exercise on metabolic outcomes are becoming increasingly apparent. We aimed to study the influence of time-restricted wheel running on whole-body energy and glucose homeostasis. Male, 8-week-old, C57BL/6NTac mice were fed either a 60% high-fat diet (HFD) or a 10% low-fat diet (LFD) for 4 weeks. Following this, mice were given access to a running wheel between zeitgeber time (ZT) 12-16 (early dark phase) or ZT 20-0 (late dark phase). Sedentary mice had access to a permanently locked wheel. Mice were housed under these conditions in metabolic chambers for 4 weeks in which LFD and HFD conditions were maintained. Following the exercise intervention, body composition and glucose tolerance were assessed. Wheel running during either the early or late dark phase resulted in metabolic improvements such as attenuation in body weight gain, enhanced glucose tolerance and reduced ectopic lipid deposition. However, late dark phase exercise resulted in a greater reduction in body weight gain, as well as enhanced metabolic flexibility and insulin sensitivity. Our data suggest that late dark phase versus early dark phase exercise confers greater metabolic adaptations in HFD-fed mice.
Collapse
Affiliation(s)
- Stephen P Ashcroft
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Amy M Ehrlich
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Krzysztof Burek
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Logan A Pendergrast
- Integrative Physiology Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Caio Y Yonamine
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Juleen R Zierath
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Integrative Physiology Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Integrative Physiology Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
3
|
Vandersmissen J, Dewachter I, Cuypers K, Hansen D. The Impact of Exercise Training on the Brain and Cognition in Type 2 Diabetes, and its Physiological Mediators: A Systematic Review. SPORTS MEDICINE - OPEN 2025; 11:42. [PMID: 40274715 PMCID: PMC12022206 DOI: 10.1186/s40798-025-00836-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 03/16/2025] [Indexed: 04/26/2025]
Abstract
BACKGROUND Type 2 diabetes (T2DM) affects brain structure and function, and is associated with an increased risk of dementia and mild cognitive impairment. It is known that exercise training has a beneficial effect on cognition and brain structure and function, at least in healthy people, but the impact of exercise training on these aspects remains to be fully elucidated in patients with T2DM. OBJECTIVE To determine the impact of exercise training on cognition and brain structure and function in T2DM, and identify the involved physiological mediators. METHODS This paper systematically reviews studies that evaluate the effect of exercise training on cognition in T2DM, and aims to indicate the most beneficial exercise modality for improving or preserving cognition in this patient group. In addition, the possible physiological mediators and targets involved in these improvements are narratively described in the second part of this review. Papers published up until the 14th of January 2025 were searched by means of the electronic databases PubMed, Embase, and Web of Science. Studies directly investigating the effect of any kind of exercise training on the brain or cognition in patients with T2DM, or animal models thereof, were included, with the exception of human studies assessing cognition only at one time point, and studies combining exercise training with other interventions (e.g. dietary changes, cognitive training, etc.). Study quality was assessed by means of the TESTEX tool for human studies, and the CAMARADES tool for animal studies. RESULTS For the systematic part of the review, 22 papers were found to be eligible. 18 out of 22 papers (81.8%) showed a significant positive effect of exercise training on cognition in T2DM, of which two studies only showed significant improvements in the minority of the cognitive tests. Four papers (18.2%) could not find a significant effect of exercise on cognition in T2DM. Resistance and endurance exercise were found to be equally effective for achieving cognitive improvement. Machine-based power training is seemingly more effective than resistance training with body weight and elastic bands to reach cognitive improvement. In addition, BDNF, lactate, leptin, adiponectin, GSK3β, GLP-1, the AMPK/SIRT1 pathway, and the PI3K/Akt pathway were identified as plausible mediators directly from studies investigating the effect of exercise training on brain structure and function in T2DM. Via these mediators, exercise training induces multiple beneficial brain changes, such as increased neuroplasticity, increased insulin sensitivity, and decreased inflammation. CONCLUSION Overall, exercise training beneficially affects cognition and brain structure and function in T2DM, with resistance and endurance exercise having similar effects. However, there is a need for additional studies, and more methodological consistency between different studies in order to define an exercise program optimal for improving cognition in T2DM. Furthermore, we were able to define several mediators involved in the effect of exercise training on cognition in T2DM, but further research is necessary to unravel the entire process.
Collapse
Affiliation(s)
- Jitske Vandersmissen
- Faculty of Rehabilitation Sciences, REVAL - Rehabilitation Research Center, Hasselt University, Wetenschapspark 7, 3590, Diepenbeek, Belgium.
| | - Ilse Dewachter
- Biomedical Research Institute, BIOMED, Hasselt University, 3590, Diepenbeek, Belgium
| | - Koen Cuypers
- Faculty of Rehabilitation Sciences, REVAL - Rehabilitation Research Center, Hasselt University, Wetenschapspark 7, 3590, Diepenbeek, Belgium
- Movement Control and Neuroplasticity Research Group, Department of Movement Sciences, Group Biomedical Sciences, KU Leuven, 3000, Leuven, Belgium
| | - Dominique Hansen
- Faculty of Rehabilitation Sciences, REVAL - Rehabilitation Research Center, Hasselt University, Wetenschapspark 7, 3590, Diepenbeek, Belgium
- Heart Centre Hasselt, Jessa Hospital, 3500, Hasselt, Belgium
| |
Collapse
|
4
|
Anderson KC, Weeldreyer NR, Leicht ZS, Angadi SS, Liu Z. Exercise Intolerance in Type 2 Diabetes: A Systematic Review and Meta-Analysis. J Am Heart Assoc 2025; 14:e035721. [PMID: 40028846 DOI: 10.1161/jaha.124.035721] [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: 03/26/2024] [Accepted: 12/18/2024] [Indexed: 03/05/2025]
Abstract
BACKGROUND To analyze the effect of type 2 diabetes (T2D) on cardiorespiratory fitness. METHODS A multilevel model using restricted maximum likelihood method was used to analyze pooled data. Inclusion criteria included adults aged >18 years, one group of individuals needed to have been diagnosed with T2D, the control group must not have had insulin resistance, the study must report peak oxygen uptake, there was no exercise training before tests of functional capacity, and subjects may not have had overt cardiovascular disease, cancer, transplant surgery, or bariatric surgery. Moderators assessed were sample demographics (age, body mass index, sex, and time since T2D diagnosis) and cardiovascular outcomes (eg, echocardiographic variables, blood pressure). RESULTS Absolute (cohorts n=30; subjects n=1152; mean difference, -0.29 L/min [95% CI, -0.37 to -0.22 L/min]; P<0.0001) and relative peak oxygen uptake (cohorts n=11; subjects n=1191; mean difference, -4.68 mL/kg per min; 95% CI, -6.94 to -2.42 mL/kg per min; P=0.001) were significantly reduced in the T2D group compared with control. Time since T2D diagnosis (β coefficient=-0.04, P=0.05) was a significant moderator of the absolute peak oxygen uptake pooled outcome. Early mitral inflow velocity/early mitral annulus velocity (β coefficient=-1.72, P=0.004) and left atrial volume index (β coefficient=-1.41, P=0.02) were significant moderators of the relative peak oxygen uptake model. CONCLUSIONS Markers of cardiac diastolic function (early mitral inflow velocity/early mitral annulus velocity and left atrial volume index) and time since diabetes diagnosis may contribute to exercise intolerance in T2D, although there is a lack of data in young/older adults and newly diagnosed individuals. As cardiorespiratory fitness predicts both all-cause mortality and cardiovascular morbidity and mortality, these data have important implications for risk reduction in individuals with T2D.
Collapse
Affiliation(s)
- Kara C Anderson
- Division of Endocrinology and Metabolism, Department of Medicine University of Virginia Health System Charlottesville VA
| | - Nathan R Weeldreyer
- Department of Kinesiology, School of Education and Human Development University of Virginia Charlottesville VA
| | - Zachary S Leicht
- Department of Kinesiology, School of Education and Human Development University of Virginia Charlottesville VA
| | - Siddhartha S Angadi
- Department of Kinesiology, School of Education and Human Development University of Virginia Charlottesville VA
| | - Zhenqi Liu
- Division of Endocrinology and Metabolism, Department of Medicine University of Virginia Health System Charlottesville VA
| |
Collapse
|
5
|
Hayes AMR, Swackhamer C, Quezada-Calvillo R, Butte NF, Sterchi EE, Nichols BL, Hamaker BR. Moderating carbohydrate digestion rate in mice promotes fat oxidation and metabolic flexibility revealed through a new approach to assess metabolic substrate utilization. Eur J Nutr 2025; 64:83. [PMID: 39904882 PMCID: PMC11908681 DOI: 10.1007/s00394-025-03585-1] [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: 06/27/2024] [Accepted: 01/08/2025] [Indexed: 02/06/2025]
Abstract
PURPOSE Superior metabolic flexibility, or the ability to efficiently switch between oxidation of carbohydrate and fat, is inversely associated with obesity and type 2 diabetes. The influence of dietary factors on metabolic flexibility is incompletely understood. This study examined the impact of dietary carbohydrate digestion rate on metabolic flexibility and metabolic substrate utilization. METHODS We employed percent relative cumulative frequency (PRCF) analyses coupled with a new application of modeling using the Mixed Weibull Cumulative Distribution function to examine respiratory exchange ratio (RER) data from adult wild-type mice and mice lacking the mucosal maltase-glucoamylase enzyme (Mgam) under different dietary carbohydrate conditions, with diets matched for total carbohydrate contents and containing different ratios of slowly digestible starch (SDS) and resistant starch (RS), or that were high in sucrose or fat. Fungal amyloglucosidase (AMG) was administered in drinking water to increase carbohydrate digestion rate. We devised a Metabolic Flexibility Factor (MFF) to quantitate metabolic flexibility for each dietary condition and mouse genotype, with higher MFF indicating higher metabolic flexibility. RESULTS Diets high in SDS exhibited lower average RER and higher metabolic flexibility (MFF) than diets high in resistant starch, sucrose, or fat. Diets containing high and intermediate amounts of SDS led to a more complete shift to fat oxidation. While mouse genotype had minimal effects on substrate oxidation and MFF, AMG supplementation shifted substrate utilization to carbohydrate oxidation and generally decreased MFF. CONCLUSIONS Consumption of slowly digestible carbohydrates improved measures of metabolic substrate utilization at the whole-body level in adult mice.
Collapse
Affiliation(s)
- Anna M R Hayes
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN, 47907, USA.
- Human and Evolutionary Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, 90089, USA.
- Department of Food Science and Technology, Oregon State University, Corvallis, OR, 97331, USA.
| | - Clay Swackhamer
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN, 47907, USA
| | - Roberto Quezada-Calvillo
- Facultad de Ciencias Quimicas, Universidad Autonoma de San Luis Potosi, Zona Universitaria,, 78210, San Luis Potosí, S.L.P., Mexico
- Department of Pediatrics, Agricultural Research Service, USDA, Children's Nutrition Research Center and Baylor College of Medicine, Houston, TX, 77030-2600, USA
| | - Nancy F Butte
- Department of Pediatrics, Agricultural Research Service, USDA, Children's Nutrition Research Center and Baylor College of Medicine, Houston, TX, 77030-2600, USA
| | - Erwin E Sterchi
- Institute of Biochemistry and Molecular Medicine, University of Bern, CH-3012, Bern, Switzerland
| | - Buford L Nichols
- Department of Pediatrics, Agricultural Research Service, USDA, Children's Nutrition Research Center and Baylor College of Medicine, Houston, TX, 77030-2600, USA.
| | - Bruce R Hamaker
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
6
|
Lecce E, Bellini A, Greco G, Martire F, Scotto di Palumbo A, Sacchetti M, Bazzucchi I. Physiological mechanisms of neuromuscular impairment in diabetes-related complications: Can physical exercise help prevent it? J Physiol 2025. [PMID: 39898972 DOI: 10.1113/jp287589] [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: 09/20/2024] [Accepted: 01/14/2025] [Indexed: 02/04/2025] Open
Abstract
Diabetes mellitus is a chronic disorder that progressively induces complications, compromising daily independence. Among these, diabetic neuropathy is particularly prevalent and contributes to substantial neuromuscular impairments in both types 1 and 2 diabetes. This condition leads to structural damage affecting both the central and peripheral nervous systems, resulting in a significant decline in sensorimotor functions. Alongside neuropathy, diabetic myopathy also contributes to muscle impairment and reduced motor performance, intensifying the neuromuscular decline. Diabetic neuropathy typically implicates neurogenic muscle atrophy, motoneuron loss and clustering of muscle fibres as a result of aberrant denervation-reinervation processes. These complications are associated with compromised neuromuscular junctions, where alterations occur in pre-synaptic vesicles, mitochondrial content and post-synaptic signalling. Neural damage is intensified by chronic hyperglycaemia and oxidative stress, exacerbating vascular dysfunction and reducing oxygen delivery. These complications imply a severe decline in neuromuscular performance, evidenced by reductions in maximal force and power output, rate of force development and muscle endurance. Furthermore, diabetes-related complications are compounded by age-related degenerative changes in long-term patients. Aerobic and resistance training offer promising approaches for managing blood glucose levels and neuromuscular function. Aerobic exercise promotes mitochondrial biogenesis and angiogenesis, supporting metabolic and cardiovascular health. Resistance training primarily enhances neural plasticity, muscle strength and hypertrophy, which are crucial factors for mitigating sarcopenia and preserving functional independence. This topical review examines current evidence on the physiological mechanisms underlying diabetic neuropathy and the potential impact of physical activity in counteracting this decline.
Collapse
Affiliation(s)
- Edoardo Lecce
- Laboratory of Exercise Physiology, Department of Movement, Human, and Health Sciences, University of 'Foro Italico', Rome, Italy
| | - Alessio Bellini
- Laboratory of Exercise Physiology, Department of Movement, Human, and Health Sciences, University of 'Foro Italico', Rome, Italy
| | - Giuseppe Greco
- Laboratory of Exercise Physiology, Department of Movement, Human, and Health Sciences, University of 'Foro Italico', Rome, Italy
| | - Fiorella Martire
- Laboratory of Exercise Physiology, Department of Movement, Human, and Health Sciences, University of 'Foro Italico', Rome, Italy
| | - Alessandro Scotto di Palumbo
- Laboratory of Exercise Physiology, Department of Movement, Human, and Health Sciences, University of 'Foro Italico', Rome, Italy
| | - Massimo Sacchetti
- Laboratory of Exercise Physiology, Department of Movement, Human, and Health Sciences, University of 'Foro Italico', Rome, Italy
| | - Ilenia Bazzucchi
- Laboratory of Exercise Physiology, Department of Movement, Human, and Health Sciences, University of 'Foro Italico', Rome, Italy
| |
Collapse
|
7
|
Harmsen J, Kotte M, Habets I, Bosschee F, Frenken K, Jorgensen JA, de Kam S, Moonen‐Kornips E, Cissen J, Doligkeit D, van de Weijer T, Erazo‐Tapia E, Buitinga M, Hoeks J, Schrauwen P. Exercise training modifies skeletal muscle clock gene expression but not 24-hour rhythmicity in substrate metabolism of men with insulin resistance. J Physiol 2024; 602:6417-6433. [PMID: 38051503 PMCID: PMC11607886 DOI: 10.1113/jp285523] [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: 08/25/2023] [Accepted: 11/14/2023] [Indexed: 12/07/2023] Open
Abstract
Twenty-four hour rhythmicity in whole-body substrate metabolism, skeletal muscle clock gene expression and mitochondrial respiration is compromised upon insulin resistance. With exercise training known to ameliorate insulin resistance, our objective was to test if exercise training can reinforce diurnal variation in whole-body and skeletal muscle metabolism in men with insulin resistance. In a single-arm longitudinal design, 10 overweight and obese men with insulin resistance performed 12 weeks of high-intensity interval training recurrently in the afternoon (between 14.00 and 18.00 h) and were tested pre- and post-exercise training, while staying in a metabolic research unit for 2 days under free-living conditions with regular meals. On the second days, indirect calorimetry was performed at 08.00, 13.00, 18.00, 23.00 and 04.00 h, muscle biopsies were taken from the vastus lateralis at 08.30, 13.30 and 23.30 h, and blood was drawn at least bi-hourly over 24 h. Participants did not lose body weight over 12 weeks, but improved body composition and exercise capacity. Exercise training resulted in reduced 24-h plasma glucose levels, but did not modify free fatty acid and triacylglycerol levels. Diurnal variation of muscle clock gene expression was modified by exercise training with period genes showing an interaction (time × exercise) effect and reduced mRNA levels at 13.00 h. Exercise training increased mitochondrial respiration without inducing diurnal variation. Twenty-four-hour substrate metabolism and energy expenditure remained unchanged. Future studies should investigate alternative exercise strategies or types of interventions (e.g. diet or drugs aiming at improving insulin sensitivity) for their capacity to reinforce diurnal variation in substrate metabolism and mitochondrial respiration. KEY POINTS: Insulin resistance is associated with blunted 24-h flexibility in whole-body substrate metabolism and skeletal muscle mitochondrial respiration, and disruptions in the skeletal muscle molecular circadian clock. We hypothesized that exercise training modifies 24-h rhythmicity in whole-body substrate metabolism and diurnal variation in skeletal muscle molecular clock and mitochondrial respiration in men with insulin resistance. We found that metabolic inflexibility over 24 h persisted after exercise training, whereas mitochondrial respiration increased independent of time of day. Gene expression of Per1-3 and Rorα in skeletal muscle changed particularly close to the time of day at which exercise training was performed. These results provide the rationale to further investigate the differential metabolic impact of differently timed exercise to treat metabolic defects of insulin resistance that manifest at a particular time of day.
Collapse
Affiliation(s)
- Jan‐Frieder Harmsen
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Marit Kotte
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Ivo Habets
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Frederieke Bosschee
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Koen Frenken
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Johanna A. Jorgensen
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Soraya de Kam
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Esther Moonen‐Kornips
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Jochem Cissen
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Daniel Doligkeit
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Tineke van de Weijer
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Edmundo Erazo‐Tapia
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Mijke Buitinga
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Joris Hoeks
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Patrick Schrauwen
- Department of Nutrition and Movement SciencesNUTRIM School for Nutrition and Translational Research in MetabolismMaastricht University Medical CenterMaastrichtThe Netherlands
| |
Collapse
|
8
|
Bhandari R, Lukas K, Lee K, Shamunee J, Almeida B, Guzman T, Echevarria M, Lindenfeld L, Nenninger C, Iukuridze A, Albanese S, Rhee JW, Chen S, Brenner C, Wong FL, Armenian SH. Feasibility of telehealth exercise and nicotinamide riboside supplementation in survivors of childhood cancer at risk for diabetes: A pilot randomized controlled trial. Pediatr Blood Cancer 2024:e31369. [PMID: 39387327 DOI: 10.1002/pbc.31369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/05/2024] [Accepted: 09/23/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Childhood cancer survivors (CCS) have a 50% higher risk of diabetes mellitus (DM) compared with the general population. Interventions in survivors with prediabetes (fasting glucose 100-125 mg/dL or hemoglobin A1c 5.7%-6.4%) may mitigate the development of DM and its attendant morbidity, but there is limited information on the feasibility of secondary prevention in this setting. METHODS This 6-week pilot feasibility 1:1 randomized controlled trial enrolled 20 CCS on a structured telehealth exercise program ± nicotinamide riboside (NR), a nicotinamide adenine dinucleotide precursor. Feasibility metrics were: (1) ≥50% of eligible CCS enrolled onto study; (2) ≥70% of participants completed baseline and end-of-study assessments; (3) ≥70% compliance with exercise and NR. Secondary endpoints included changes in biomarkers associated with glucose homeostasis and muscle health. RESULTS Median age (years) at cancer diagnosis was 16.5 (range, 1.5-21.5) and 35.5 (range, 18.0-67.0) at study enrollment. Enrollment rate was 87%, and 85% of participants completed baseline and end-of-study assessments. The mean percentage of exercise sessions completed was 86.6%; NR compliance was > 90%. There were no severe adverse events attributable to study interventions. Secondary endpoints were not significantly different between study arms at study completion. Myostatin decrease was observed in participants who completed a higher median number of exercise sessions and was associated with decreased intramuscular adipose tissue and increased lower extremity muscle cross-sectional area. CONCLUSIONS A telehealth exercise intervention ± NR supplementation was feasible in CCS with prediabetes. Future studies in larger cohorts may be needed to evaluate their beneficial effects on muscle health and DM risk among CCS.
Collapse
Affiliation(s)
- Rusha Bhandari
- Department of Population Sciences, City of Hope, Duarte, California, USA
- Department of Pediatrics, City of Hope, Duarte, California, USA
| | - Kara Lukas
- Department of Population Sciences, City of Hope, Duarte, California, USA
| | - Kyuwan Lee
- Department of Population Sciences, City of Hope, Duarte, California, USA
| | - Justin Shamunee
- Department of Population Sciences, City of Hope, Duarte, California, USA
| | - Brady Almeida
- Department of Population Sciences, City of Hope, Duarte, California, USA
| | - Tati Guzman
- Department of Population Sciences, City of Hope, Duarte, California, USA
| | - Meagan Echevarria
- Department of Population Sciences, City of Hope, Duarte, California, USA
| | - Lanie Lindenfeld
- Department of Population Sciences, City of Hope, Duarte, California, USA
| | | | - Aleksi Iukuridze
- Department of Population Sciences, City of Hope, Duarte, California, USA
| | - Sophia Albanese
- Department of Population Sciences, City of Hope, Duarte, California, USA
| | - June-Wha Rhee
- Department of Medicine, City of Hope, Duarte, California, USA
| | - Sitong Chen
- Department of Population Sciences, City of Hope, Duarte, California, USA
| | - Charles Brenner
- Department of Diabetes and Cancer Metabolism, City of Hope, Duarte, California, USA
| | - F Lennie Wong
- Department of Population Sciences, City of Hope, Duarte, California, USA
| | - Saro H Armenian
- Department of Population Sciences, City of Hope, Duarte, California, USA
- Department of Pediatrics, City of Hope, Duarte, California, USA
| |
Collapse
|
9
|
Latva-Rasku A, Rebelos E, Tuisku J, Aarnio R, Bhowmik A, Keskinen H, Laurila S, Lahesmaa-Hatting M, Pekkarinen L, Isackson H, Kirjavainen AK, Koffert J, Heurling K, Nummenmaa L, Ferrannini E, Oldgren J, Oscarsson J, Nuutila P. SGLT2 Inhibitor Dapagliflozin Increases Skeletal Muscle and Brain Fatty Acid Uptake in Individuals With Type 2 Diabetes: A Randomized Double-Blind Placebo-Controlled Positron Emission Tomography Study. Diabetes Care 2024; 47:1630-1637. [PMID: 38941156 DOI: 10.2337/dc24-0470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/10/2024] [Indexed: 06/30/2024]
Abstract
OBJECTIVE The aim of this study was to investigate the impact of the sodium-glucose cotransporter 2 (SGLT2) inhibitor dapagliflozin on tissue fatty acid (FA) uptake in the skeletal muscle, brain, small intestine, and subcutaneous and visceral adipose tissue of individuals with type 2 diabetes by using positron emission tomography (PET). RESEARCH DESIGN AND METHODS In a 6-week randomized double-blind placebo-controlled trial, 53 patients with type 2 diabetes treated with metformin received either 10 mg dapagliflozin or placebo daily. Tissue FA uptake was quantified at baseline and end of treatment with PET and the long-chain FA analog radiotracer 14(R,S)-[18F]fluoro-6-thia-heptadecanoic acid. Treatment effects were assessed using ANCOVA, and the results are reported as least square means and 95% CIs for the difference between groups. RESULTS A total of 38 patients (dapagliflozin n = 21; placebo n = 17) completed the study. After 6 weeks, skeletal muscle FA uptake was increased by dapagliflozin compared with placebo (1.0 [0.07, 2.0] μmol ⋅ 100 g-1 ⋅ min-1; P = 0.032), whereas uptake was not significantly changed in the small intestine or visceral or subcutaneous adipose tissue. Dapagliflozin treatment significantly increased whole-brain FA uptake (0.10 [0.02, 0.17] μmol ⋅ 100 g-1 ⋅ min-1; P = 0.01), an effect observed in both gray and white matter regions. CONCLUSIONS Six weeks of treatment with dapagliflozin increases skeletal muscle and brain FA uptake, partly driven by a rise in free FA availability. This finding is in accordance with previous indirect measurements showing enhanced FA metabolism in response to SGLT2 inhibition and extends the notion of a shift toward increased FA use to muscle and brain.
Collapse
Affiliation(s)
- Aino Latva-Rasku
- Turku PET Centre, University of Turku, Turku, Finland
- Department of Endocrinology, Turku University Hospital, Turku, Finland
| | - Eleni Rebelos
- Turku PET Centre, University of Turku, Turku, Finland
| | - Jouni Tuisku
- Turku PET Centre, University of Turku, Turku, Finland
| | | | - Achol Bhowmik
- Turku PET Centre, University of Turku, Turku, Finland
| | | | - Sanna Laurila
- Turku PET Centre, University of Turku, Turku, Finland
- Heart Center, Turku University Hospital, Turku, Finland
| | | | - Laura Pekkarinen
- Turku PET Centre, University of Turku, Turku, Finland
- Department of Endocrinology, Turku University Hospital, Turku, Finland
| | - Henrik Isackson
- Clinical Physiology and Cardiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Integrative Physiology, Medical Cell Biology, Uppsala University Hospital, Uppsala, Sweden
| | - Anna K Kirjavainen
- Radiopharmaceutical Chemistry Laboratory, Turku PET Centre, University of Turku, Turku, Finland
| | - Jukka Koffert
- Turku PET Centre, University of Turku, Turku, Finland
| | | | - Lauri Nummenmaa
- Turku PET Centre, University of Turku, Turku, Finland
- Department of Psychology, University of Turku, Turku, Finland
| | - Ele Ferrannini
- National Research Council Institute of Clinical Physiology, Pisa, Italy
| | - Jonas Oldgren
- Clinical Physiology and Cardiology, Department of Medical Sciences, Uppsala University, Uppsala, Sweden
- Uppsala Clinical Research Center, Uppsala University, Uppsala, Sweden
| | - Jan Oscarsson
- Late-Stage Development, Cardiovascular, Renal, and Metabolism, BioPharmaceuticals Research and Development, AstraZeneca, Gothenburg, Sweden
| | - Pirjo Nuutila
- Turku PET Centre, University of Turku, Turku, Finland
- Department of Endocrinology, Turku University Hospital, Turku, Finland
| |
Collapse
|
10
|
Esteves JV, Stanford KI. Exercise as a tool to mitigate metabolic disease. Am J Physiol Cell Physiol 2024; 327:C587-C598. [PMID: 38981607 PMCID: PMC11427015 DOI: 10.1152/ajpcell.00144.2024] [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: 03/12/2024] [Revised: 06/28/2024] [Accepted: 06/28/2024] [Indexed: 07/11/2024]
Abstract
Metabolic diseases, notably obesity and type 2 diabetes (T2D), have reached alarming proportions and constitute a significant global health challenge, emphasizing the urgent need for effective preventive and therapeutic strategies. In contrast, exercise training emerges as a potent intervention, exerting numerous positive effects on metabolic health through adaptations to the metabolic tissues. Here, we reviewed the major features of our current understanding with respect to the intricate interplay between metabolic diseases and key metabolic tissues, including adipose tissue, skeletal muscle, and liver, describing some of the main underlying mechanisms driving pathogenesis, as well as the role of exercise to combat and treat obesity and metabolic disease.
Collapse
Affiliation(s)
- Joao Victor Esteves
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
- Division of General and Gastrointestinal Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| | - Kristin I Stanford
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
- Division of General and Gastrointestinal Surgery, Department of Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio, United States
| |
Collapse
|
11
|
Curl CC, Leija RG, Arevalo JA, Osmond AD, Duong JJ, Huie MJ, Masharani U, Horning MA, Brooks GA. Altered glucose kinetics occurs with aging: a new outlook on metabolic flexibility. Am J Physiol Endocrinol Metab 2024; 327:E217-E228. [PMID: 38895979 PMCID: PMC11427093 DOI: 10.1152/ajpendo.00091.2024] [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: 02/27/2024] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 06/21/2024]
Abstract
Our purpose was to determine how age affects metabolic flexibility and underlying glucose kinetics in healthy young and older adults. Therefore, glucose and lactate tracers along with pulmonary gas exchange data were used to determine glucose kinetics and respiratory exchange ratios [RER = carbon dioxide production (V̇co2)/oxygen consumption (V̇o2)] during a 2-h 75-g oral glucose tolerance test (OGTT). After an 12-h overnight fast, 28 participants, 15 young (21-35 yr; 7 men and 8 women) and 13 older (60-80 yr; 7 men and 6 women), received venous primed-continuous infusions of [6,6-2H]glucose and [3-13C]lactate with a [Formula: see text] bolus. After a 90-min metabolic stabilization and tracer equilibration period, volunteers underwent an OGTT. Arterialized glucose concentrations ([glucose]) started to rise 15 min post glucose consumption, peaked at 60 min, and remained elevated. As assessed by rates of appearance (Ra) and disposal (Rd) and metabolic clearance rate (MCR), glucose kinetics were suppressed in older compared to young individuals. As well, unlike in young individuals, fractional gluconeogenesis (fGNG) remained elevated in the older population after the oral glucose challenge. Finally, there were no differences in 12-h fasting baseline or peak RER values following an oral glucose challenge in older compared to young men and women, making RER an incomplete measure of metabolic flexibility in the volunteers we evaluated. Our study revealed that glucose kinetics are significantly altered in a healthy aged population after a glucose challenge. Furthermore, those physiological deficits are not detected from changes in RER during an OGTT.NEW & NOTEWORTHY To determine metabolic flexibility in response to an OGTT, we studied healthy young and older men and women to determine glucose kinetics and changes in RER. Compared to young subjects, glucose kinetics were suppressed in older healthy individuals during an OGTT. Surprisingly, the age-related changes in glucose flux were not reflected in RER measurements; thus, RER measurements do not give a complete view of metabolic flexibility in healthy individuals.
Collapse
Affiliation(s)
- Casey C Curl
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, California, United States
| | - Robert G Leija
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, California, United States
| | - Jose A Arevalo
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, California, United States
| | - Adam D Osmond
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, California, United States
| | - Justin J Duong
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, California, United States
| | - Melvin J Huie
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, California, United States
| | - Umesh Masharani
- Department of Endocrinology, School of Medicine, University of California, San Francisco, California, United States
| | - Michael A Horning
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, California, United States
| | - George A Brooks
- Exercise Physiology Laboratory, Department of Integrative Biology, University of California, Berkeley, California, United States
| |
Collapse
|
12
|
Park S, Cha HN, Shin MG, Park S, Kim Y, Kim MS, Shin KH, Thoudam T, Lee EJ, Wolfe RR, Dan J, Koh JH, Kim IY, Choi I, Lee IK, Sung HK, Park SY. Inhibitory Regulation of FOXO1 in PPARδ Expression Drives Mitochondrial Dysfunction and Insulin Resistance. Diabetes 2024; 73:1084-1098. [PMID: 38656552 DOI: 10.2337/db23-0432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 04/09/2024] [Indexed: 04/26/2024]
Abstract
Forkhead box O1 (FOXO1) regulates muscle growth, but the metabolic role of FOXO1 in skeletal muscle and its mechanisms remain unclear. To explore the metabolic role of FOXO1 in skeletal muscle, we generated skeletal muscle-specific Foxo1 inducible knockout (mFOXO1 iKO) mice and fed them a high-fat diet to induce obesity. We measured insulin sensitivity, fatty acid oxidation, mitochondrial function, and exercise capacity in obese mFOXO1 iKO mice and assessed the correlation between FOXO1 and mitochondria-related protein in the skeletal muscle of patients with diabetes. Obese mFOXO1 iKO mice exhibited improved mitochondrial respiratory capacity, which was followed by attenuated insulin resistance, enhanced fatty acid oxidation, and improved skeletal muscle exercise capacity. Transcriptional inhibition of FOXO1 in peroxisome proliferator-activated receptor δ (PPARδ) expression was confirmed in skeletal muscle, and deletion of PPARδ abolished the beneficial effects of FOXO1 deficiency. FOXO1 protein levels were higher in the skeletal muscle of patients with diabetes and negatively correlated with PPARδ and electron transport chain protein levels. These findings highlight FOXO1 as a new repressor in PPARδ gene expression in skeletal muscle and suggest that FOXO1 links insulin resistance and mitochondrial dysfunction in skeletal muscle via PPARδ. ARTICLE HIGHLIGHTS
Collapse
Affiliation(s)
- Soyoung Park
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Republic of Korea
- Senotherapy-Based Metabolic Diseases Control Research Center, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Hye-Na Cha
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Republic of Korea
- Senotherapy-Based Metabolic Diseases Control Research Center, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Min-Gyeong Shin
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| | - Sanghee Park
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Yeongmin Kim
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology, Gachon University, Incheon, Republic of Korea
| | - Min-Seob Kim
- Department of Fundamental Environment Research, Environmental Measurement and Analysis Center, National Institute of Environmental Research, Incheon, Republic of Korea
| | - Kyung-Hoon Shin
- Department of Marine Sciences and Convergent Technology, Hanyang University, Ansan, Republic of Korea
| | - Themis Thoudam
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea
| | - Eun Ju Lee
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - Robert R Wolfe
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Jinmyoung Dan
- Department of Orthopedic Surgery, College of Medicine, CHA University, Gumi, Republic of Korea
| | - Jin-Ho Koh
- Department of Convergence Medicine, Wonju College of Medicine, Yonsei University, Wonju, Republic of Korea
| | - Il-Young Kim
- Department of Molecular Medicine, College of Medicine, Gachon University, Incheon, Republic of Korea
| | - Inho Choi
- Department of Medical Biotechnology, Yeungnam University, Gyeongsan, Republic of Korea
| | - In-Kyu Lee
- Research Institute of Aging and Metabolism, Kyungpook National University, Daegu, Republic of Korea
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Kyungpook National University Hospital, Daegu, Republic of Korea
| | - Hoon-Ki Sung
- The Hospital for Sick Children Research Institute & Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - So-Young Park
- Department of Physiology, College of Medicine, Yeungnam University, Daegu, Republic of Korea
- Senotherapy-Based Metabolic Diseases Control Research Center, College of Medicine, Yeungnam University, Daegu, Republic of Korea
| |
Collapse
|
13
|
Poljšak B, Milisav I. Decreasing Intracellular Entropy by Increasing Mitochondrial Efficiency and Reducing ROS Formation-The Effect on the Ageing Process and Age-Related Damage. Int J Mol Sci 2024; 25:6321. [PMID: 38928027 PMCID: PMC11203720 DOI: 10.3390/ijms25126321] [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: 04/23/2024] [Revised: 06/01/2024] [Accepted: 06/04/2024] [Indexed: 06/28/2024] Open
Abstract
A hypothesis is presented to explain how the ageing process might be influenced by optimizing mitochondrial efficiency to reduce intracellular entropy. Research-based quantifications of entropy are scarce. Non-equilibrium metabolic reactions and compartmentalization were found to contribute most to lowering entropy in the cells. Like the cells, mitochondria are thermodynamically open systems exchanging matter and energy with their surroundings-the rest of the cell. Based on the calculations from cancer cells, glycolysis was reported to produce less entropy than mitochondrial oxidative phosphorylation. However, these estimations depended on the CO2 concentration so that at slightly increased CO2, it was oxidative phosphorylation that produced less entropy. Also, the thermodynamic efficiency of mitochondrial respiratory complexes varies depending on the respiratory state and oxidant/antioxidant balance. Therefore, in spite of long-standing theoretical and practical efforts, more measurements, also in isolated mitochondria, with intact and suboptimal respiration, are needed to resolve the issue. Entropy increases in ageing while mitochondrial efficiency of energy conversion, quality control, and turnover mechanisms deteriorate. Optimally functioning mitochondria are necessary to meet energy demands for cellular defence and repair processes to attenuate ageing. The intuitive approach of simply supplying more metabolic fuels (more nutrients) often has the opposite effect, namely a decrease in energy production in the case of nutrient overload. Excessive nutrient intake and obesity accelerate ageing, while calorie restriction without malnutrition can prolong life. Balanced nutrient intake adapted to needs/activity-based high ATP requirement increases mitochondrial respiratory efficiency and leads to multiple alterations in gene expression and metabolic adaptations. Therefore, rather than overfeeding, it is necessary to fine-tune energy production by optimizing mitochondrial function and reducing oxidative stress; the evidence is discussed in this paper.
Collapse
Affiliation(s)
- Borut Poljšak
- Laboratory of Oxidative Stress Research, Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, SI-1000 Ljubljana, Slovenia;
| | - Irina Milisav
- Laboratory of Oxidative Stress Research, Faculty of Health Sciences, University of Ljubljana, Zdravstvena pot 5, SI-1000 Ljubljana, Slovenia;
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Zaloska 4, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
14
|
Espino-Gonzalez E, Dalbram E, Mounier R, Gondin J, Farup J, Jessen N, Treebak JT. Impaired skeletal muscle regeneration in diabetes: From cellular and molecular mechanisms to novel treatments. Cell Metab 2024; 36:1204-1236. [PMID: 38490209 DOI: 10.1016/j.cmet.2024.02.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 01/10/2024] [Accepted: 02/22/2024] [Indexed: 03/17/2024]
Abstract
Diabetes represents a major public health concern with a considerable impact on human life and healthcare expenditures. It is now well established that diabetes is characterized by a severe skeletal muscle pathology that limits functional capacity and quality of life. Increasing evidence indicates that diabetes is also one of the most prevalent disorders characterized by impaired skeletal muscle regeneration, yet underlying mechanisms and therapeutic treatments remain poorly established. In this review, we describe the cellular and molecular alterations currently known to occur during skeletal muscle regeneration in people with diabetes and animal models of diabetes, including its associated comorbidities, e.g., obesity, hyperinsulinemia, and insulin resistance. We describe the role of myogenic and non-myogenic cell types on muscle regeneration in conditions with or without diabetes. Therapies for skeletal muscle regeneration and gaps in our knowledge are also discussed, while proposing future directions for the field.
Collapse
Affiliation(s)
- Ever Espino-Gonzalez
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Emilie Dalbram
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark
| | - Rémi Mounier
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - Julien Gondin
- Institut NeuroMyoGène, Unité Physiopathologie et Génétique du Neurone et du Muscle, Université Claude Bernard Lyon 1, CNRS UMR 5261, Inserm U1315, Univ Lyon, Lyon, France
| | - Jean Farup
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Niels Jessen
- Department of Biomedicine, Aarhus University, Aarhus 8000, Denmark; Steno Diabetes Center Aarhus, Aarhus University Hospital, Aarhus 8200, Denmark; Department of Clinical Pharmacology, Aarhus University Hospital, Aarhus 8200, Denmark
| | - Jonas T Treebak
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen 2200, Denmark.
| |
Collapse
|
15
|
Flores-Opazo M, Kopinke D, Helmbacher F, Fernández-Verdejo R, Tuñón-Suárez M, Lynch GS, Contreras O. Fibro-adipogenic progenitors in physiological adipogenesis and intermuscular adipose tissue remodeling. Mol Aspects Med 2024; 97:101277. [PMID: 38788527 PMCID: PMC11692456 DOI: 10.1016/j.mam.2024.101277] [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: 02/01/2024] [Revised: 04/27/2024] [Accepted: 05/13/2024] [Indexed: 05/26/2024]
Abstract
Excessive accumulation of intermuscular adipose tissue (IMAT) is a common pathological feature in various metabolic and health conditions and can cause muscle atrophy, reduced function, inflammation, insulin resistance, cardiovascular issues, and unhealthy aging. Although IMAT results from fat accumulation in muscle, the mechanisms underlying its onset, development, cellular components, and functions remain unclear. IMAT levels are influenced by several factors, such as changes in the tissue environment, muscle type and origin, extent and duration of trauma, and persistent activation of fibro-adipogenic progenitors (FAPs). FAPs are a diverse and transcriptionally heterogeneous population of stromal cells essential for tissue maintenance, neuromuscular stability, and tissue regeneration. However, in cases of chronic inflammation and pathological conditions, FAPs expand and differentiate into adipocytes, resulting in the development of abnormal and ectopic IMAT. This review discusses the role of FAPs in adipogenesis and how they remodel IMAT. It highlights evidence supporting FAPs and FAP-derived adipocytes as constituents of IMAT, emphasizing their significance in adipose tissue maintenance and development, as well as their involvement in metabolic disorders, chronic pathologies and diseases. We also investigated the intricate molecular pathways and cell interactions governing FAP behavior, adipogenesis, and IMAT accumulation in chronic diseases and muscle deconditioning. Finally, we hypothesize that impaired cellular metabolic flexibility in dysfunctional muscles impacts FAPs, leading to IMAT. A deeper understanding of the biology of IMAT accumulation and the mechanisms regulating FAP behavior and fate are essential for the development of new therapeutic strategies for several debilitating conditions.
Collapse
Affiliation(s)
| | - Daniel Kopinke
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, 32610, FL, USA; Myology Institute, University of Florida College of Medicine, Gainesville, FL, USA.
| | | | - Rodrigo Fernández-Verdejo
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA; Laboratorio de Fisiología Del Ejercicio y Metabolismo (LABFEM), Escuela de Kinesiología, Facultad de Medicina, Universidad Finis Terrae, Chile.
| | - Mauro Tuñón-Suárez
- Laboratorio de Fisiología Del Ejercicio y Metabolismo (LABFEM), Escuela de Kinesiología, Facultad de Medicina, Universidad Finis Terrae, Chile.
| | - Gordon S Lynch
- Centre for Muscle Research, Department of Anatomy and Physiology, The University of Melbourne, Melbourne, Victoria, Parkville 3010, Australia.
| | - Osvaldo Contreras
- Developmental and Regenerative Biology Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW, 2010, Australia; School of Clinical Medicine, UNSW Sydney, Kensington, NSW 2052, Australia.
| |
Collapse
|
16
|
Garneau L, Mulvihill EE, Smith SR, Sparks LM, Aguer C. Myokine Secretion following an Aerobic Exercise Intervention in Individuals with Type 2 Diabetes with or without Exercise Resistance. Int J Mol Sci 2024; 25:4889. [PMID: 38732106 PMCID: PMC11084395 DOI: 10.3390/ijms25094889] [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: 03/22/2024] [Revised: 04/19/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Type 2 diabetes (T2D) is characterized by muscle metabolic dysfunction that exercise can minimize, but some patients do not respond to an exercise intervention. Myokine secretion is intrinsically altered in patients with T2D, but the role of myokines in exercise resistance in this patient population has never been studied. We sought to determine if changes in myokine secretion were linked to the response to an exercise intervention in patients with T2D. The participants followed a 10-week aerobic exercise training intervention, and patients with T2D were grouped based on muscle mitochondrial function improvement (responders versus non-responders). We measured myokines in serum and cell-culture medium of myotubes derived from participants pre- and post-intervention and in response to an in vitro model of muscle contraction. We also quantified the expression of genes related to inflammation in the myotubes pre- and post-intervention. No significant differences were detected depending on T2D status or response to exercise in the biological markers measured, with the exception of modest differences in expression patterns for certain myokines (IL-1β, IL-8, IL-10, and IL-15). Further investigation into the molecular mechanisms involving myokines may explain exercise resistance with T2D; however, the role in metabolic adaptations to exercise in T2D requires further investigation.
Collapse
Affiliation(s)
- Léa Garneau
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (L.G.); (E.E.M.)
- Institut du Savoir Montfort, Ottawa, ON K1K 0T2, Canada
| | - Erin E. Mulvihill
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (L.G.); (E.E.M.)
- University of Ottawa Heart Institute, Ottawa, ON K1Y 4W7, Canada
| | - Steven R. Smith
- Translational Research Institute for Metabolism and Diabetes, AdventHealth Orlando, Orlando, FL 32804, USA; (S.R.S.); (L.M.S.)
| | - Lauren M. Sparks
- Translational Research Institute for Metabolism and Diabetes, AdventHealth Orlando, Orlando, FL 32804, USA; (S.R.S.); (L.M.S.)
| | - Céline Aguer
- Faculty of Medicine, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON K1H 8M5, Canada; (L.G.); (E.E.M.)
- Institut du Savoir Montfort, Ottawa, ON K1K 0T2, Canada
- Faculty of Medicine and Health Sciences, Department of Physiology, McGill University–Campus Outaouais, Gatineau, QC J8V 3T4, Canada
- Faculty of Health Sciences, School of Human Kinetics, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
17
|
Ashcroft SP, Stocks B, Egan B, Zierath JR. Exercise induces tissue-specific adaptations to enhance cardiometabolic health. Cell Metab 2024; 36:278-300. [PMID: 38183980 DOI: 10.1016/j.cmet.2023.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/06/2023] [Accepted: 12/05/2023] [Indexed: 01/08/2024]
Abstract
The risk associated with multiple cancers, cardiovascular disease, diabetes, and all-cause mortality is decreased in individuals who meet the current recommendations for physical activity. Therefore, regular exercise remains a cornerstone in the prevention and treatment of non-communicable diseases. An acute bout of exercise results in the coordinated interaction between multiple tissues to meet the increased energy demand of exercise. Over time, the associated metabolic stress of each individual exercise bout provides the basis for long-term adaptations across tissues, including the cardiovascular system, skeletal muscle, adipose tissue, liver, pancreas, gut, and brain. Therefore, regular exercise is associated with a plethora of benefits throughout the whole body, including improved cardiorespiratory fitness, physical function, and glycemic control. Overall, we summarize the exercise-induced adaptations that occur within multiple tissues and how they converge to ultimately improve cardiometabolic health.
Collapse
Affiliation(s)
- Stephen P Ashcroft
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ben Stocks
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Brendan Egan
- School of Health and Human Performance, Dublin City University, Dublin, Ireland
| | - Juleen R Zierath
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Integrative Physiology, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden; Integrative Physiology, Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
18
|
Bartlett MF, Fitzgerald LF, Nagarajan R, Kent JA. Measurements of in vivo skeletal muscle oxidative capacity are lower following sustained isometric compared with dynamic contractions. Appl Physiol Nutr Metab 2024; 49:250-264. [PMID: 37906958 DOI: 10.1139/apnm-2023-0315] [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] [Indexed: 11/02/2023]
Abstract
Human skeletal muscle oxidative capacity can be quantified non-invasively using 31-phosphorus magnetic resonance spectroscopy (31P-MRS) to measure the rate constant of phosphocreatine (PCr) recovery (kPCr) following contractions. In the quadricep muscles, several studies have quantified kPCr following 24-30 s of sustained maximal voluntary isometric contraction (MVIC). This approach has the advantage of simplicity but is potentially problematic because sustained MVICs inhibit perfusion, which may limit muscle oxygen availability or increase the intracellular metabolic perturbation, and thus affect kPCr. Alternatively, dynamic contractions allow reperfusion between contractions, which may avoid limitations in oxygen delivery. To determine whether dynamic contraction protocols elicit greater kPCr than sustained MVIC protocols, we used a cross-sectional design to compare quadriceps kPCr in 22 young and 11 older healthy adults following 24 s of maximal voluntary: (1) sustained MVIC and (2) dynamic (MVDC; 120°·s-1, 1 every 2 s) contractions. Muscle kPCr was ∼20% lower following the MVIC protocol compared with the MVDC protocol (p ≤ 0.001), though this was less evident in older adults (p = 0.073). Changes in skeletal muscle pH (p ≤ 0.001) and PME accumulation (p ≤ 0.001) were greater following the sustained MVIC protocol, and pH (p ≤ 0.001) and PME (p ≤ 0.001) recovery were slower. These results demonstrate that (i) a brief, sustained MVIC yields a lower value for skeletal muscle oxidative capacity than an MVDC protocol of similar duration and (ii) this difference may not be consistent across populations (e.g., young vs. old). Thus, the potential effect of contraction protocol on comparisons of kPCr in different study groups requires careful consideration in the future.
Collapse
Affiliation(s)
- Miles F Bartlett
- Department of KinesiologyMuscle Physiology Laboratory, University of Massachusetts Amherst, MA 01003, USA
| | - Liam F Fitzgerald
- Department of KinesiologyMuscle Physiology Laboratory, University of Massachusetts Amherst, MA 01003, USA
| | - Rajakumar Nagarajan
- Human Magnetic Resonance Center, Institute for Applied Life Sciences (IALS), University of Massachusetts Amherst, MA 01003, USA
| | - Jane A Kent
- Department of KinesiologyMuscle Physiology Laboratory, University of Massachusetts Amherst, MA 01003, USA
| |
Collapse
|
19
|
Gonzalez-Armenta JL, Bergstrom J, Lee J, Furdui CM, Nicklas BJ, Molina AJA. Serum factors mediate changes in mitochondrial bioenergetics associated with diet and exercise interventions. GeroScience 2024; 46:349-365. [PMID: 37368157 PMCID: PMC10828137 DOI: 10.1007/s11357-023-00855-w] [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: 04/27/2023] [Accepted: 06/15/2023] [Indexed: 06/28/2023] Open
Abstract
Mitochondrial improvements resulting from behavioral interventions, such as diet and exercise, are systemic and apparent across multiple tissues. Here, we test the hypothesis that factors present in serum, and therefore circulating throughout the body, can mediate changes in mitochondrial function in response to intervention. To investigate this, we used stored serum from a clinical trial comparing resistance training (RT) and RT plus caloric restriction (RT + CR) to examine effects of blood borne circulating factors on myoblasts in vitro. We report that exposure to dilute serum is sufficient to mediate bioenergetic benefits of these interventions. Additionally, serum-mediated bioenergetic changes can differentiate between interventions, recapitulate sex differences in bioenergetic responses, and is linked to improvements in physical function and inflammation. Using metabolomics, we identified circulating factors associated with changes in mitochondrial bioenergetics and the effects of interventions. This study provides new evidence that circulating factors play a role in the beneficial effects of interventions that improve healthspan among older adults. Understanding the factors that drive improvements in mitochondrial function is a key step towards predicting intervention outcomes and developing strategies to countermand systemic age-related bioenergetic decline.
Collapse
Affiliation(s)
- Jenny L Gonzalez-Armenta
- Section On Gerontology and Geriatrics, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Jaclyn Bergstrom
- Division of Geriatrics, Gerontology, and Palliative Care, Department of Medicine, University of California San Diego School of Medicine, 9500 Gilman Drive, MC 0665, La Jolla, CA, 92093-0665, USA
| | - Jingyun Lee
- Proteomics and Metabolomics Shared Resource, Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Cristina M Furdui
- Section On Molecular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Barbara J Nicklas
- Section On Gerontology and Geriatrics, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - Anthony J A Molina
- Division of Geriatrics, Gerontology, and Palliative Care, Department of Medicine, University of California San Diego School of Medicine, 9500 Gilman Drive, MC 0665, La Jolla, CA, 92093-0665, USA.
| |
Collapse
|
20
|
Billon C, Schoepke E, Avdagic A, Chatterjee A, Butler AA, Elgendy B, Walker JK, Burris TP. A Synthetic ERR Agonist Alleviates Metabolic Syndrome. J Pharmacol Exp Ther 2024; 388:232-240. [PMID: 37739806 PMCID: PMC10801787 DOI: 10.1124/jpet.123.001733] [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: 05/09/2023] [Revised: 07/27/2023] [Accepted: 08/07/2022] [Indexed: 09/24/2023] Open
Abstract
Physical exercise induces physiologic adaptations and is effective at reducing the risk of premature death from all causes. Pharmacological exercise mimetics may be effective in the treatment of a range of diseases including obesity and metabolic syndrome. Previously, we described the development of SLU-PP-332, an agonist for the estrogen-related receptor (ERR)α, β, and γ nuclear receptors that activates an acute aerobic exercise program. Here we examine the effects of this exercise mimetic in mouse models of obesity and metabolic syndrome. Diet-induced obese or ob/ob mice were administered SLU-PP-332, and the effects on a range of metabolic parameters were assessed. SLU-PP-332 administration mimics exercise-induced benefits on whole-body metabolism in mice including increased energy expenditure and fatty acid oxidation. These effects were accompanied by decreased fat mass accumulation. Additionally, the ERR agonist effectively reduced obesity and improved insulin sensitivity in models of metabolic syndrome. Pharmacological activation of ERR may be an effective method to treat metabolic syndrome and obesity. SIGNIFICANCE STATEMENT: An estrogen receptor-related orphan receptor agonist, SLU-PP-332, with exercise mimetic activity, holds promise as a therapeutic to treat metabolic diseases by decreasing fat mass in mouse models of obesity.
Collapse
Affiliation(s)
- Cyrielle Billon
- Center for Clinical Pharmacology, St. Louis College of Pharmacy, Washington University School of Medicine, University of Health Sciences and Pharmacy, St. Louis, Missouri (C.B., A.A., B.E.); Department of Pharmacology & Physiology (E.S., A.C., A.A.B., J.K.W.) and Department of Chemistry (J.K.W.), Saint Louis University School of Medicine, St. Louis, Missouri; and University of Florida Genetics Institute, Gainesville, Florida (T.P.B.)
| | - Emmalie Schoepke
- Center for Clinical Pharmacology, St. Louis College of Pharmacy, Washington University School of Medicine, University of Health Sciences and Pharmacy, St. Louis, Missouri (C.B., A.A., B.E.); Department of Pharmacology & Physiology (E.S., A.C., A.A.B., J.K.W.) and Department of Chemistry (J.K.W.), Saint Louis University School of Medicine, St. Louis, Missouri; and University of Florida Genetics Institute, Gainesville, Florida (T.P.B.)
| | - Amer Avdagic
- Center for Clinical Pharmacology, St. Louis College of Pharmacy, Washington University School of Medicine, University of Health Sciences and Pharmacy, St. Louis, Missouri (C.B., A.A., B.E.); Department of Pharmacology & Physiology (E.S., A.C., A.A.B., J.K.W.) and Department of Chemistry (J.K.W.), Saint Louis University School of Medicine, St. Louis, Missouri; and University of Florida Genetics Institute, Gainesville, Florida (T.P.B.)
| | - Arindam Chatterjee
- Center for Clinical Pharmacology, St. Louis College of Pharmacy, Washington University School of Medicine, University of Health Sciences and Pharmacy, St. Louis, Missouri (C.B., A.A., B.E.); Department of Pharmacology & Physiology (E.S., A.C., A.A.B., J.K.W.) and Department of Chemistry (J.K.W.), Saint Louis University School of Medicine, St. Louis, Missouri; and University of Florida Genetics Institute, Gainesville, Florida (T.P.B.)
| | - Andrew A Butler
- Center for Clinical Pharmacology, St. Louis College of Pharmacy, Washington University School of Medicine, University of Health Sciences and Pharmacy, St. Louis, Missouri (C.B., A.A., B.E.); Department of Pharmacology & Physiology (E.S., A.C., A.A.B., J.K.W.) and Department of Chemistry (J.K.W.), Saint Louis University School of Medicine, St. Louis, Missouri; and University of Florida Genetics Institute, Gainesville, Florida (T.P.B.)
| | - Bahaa Elgendy
- Center for Clinical Pharmacology, St. Louis College of Pharmacy, Washington University School of Medicine, University of Health Sciences and Pharmacy, St. Louis, Missouri (C.B., A.A., B.E.); Department of Pharmacology & Physiology (E.S., A.C., A.A.B., J.K.W.) and Department of Chemistry (J.K.W.), Saint Louis University School of Medicine, St. Louis, Missouri; and University of Florida Genetics Institute, Gainesville, Florida (T.P.B.)
| | - John K Walker
- Center for Clinical Pharmacology, St. Louis College of Pharmacy, Washington University School of Medicine, University of Health Sciences and Pharmacy, St. Louis, Missouri (C.B., A.A., B.E.); Department of Pharmacology & Physiology (E.S., A.C., A.A.B., J.K.W.) and Department of Chemistry (J.K.W.), Saint Louis University School of Medicine, St. Louis, Missouri; and University of Florida Genetics Institute, Gainesville, Florida (T.P.B.)
| | - Thomas P Burris
- Center for Clinical Pharmacology, St. Louis College of Pharmacy, Washington University School of Medicine, University of Health Sciences and Pharmacy, St. Louis, Missouri (C.B., A.A., B.E.); Department of Pharmacology & Physiology (E.S., A.C., A.A.B., J.K.W.) and Department of Chemistry (J.K.W.), Saint Louis University School of Medicine, St. Louis, Missouri; and University of Florida Genetics Institute, Gainesville, Florida (T.P.B.)
| |
Collapse
|
21
|
Pataky MW, Kumar AP, Gaul DA, Moore SG, Dasari S, Robinson MM, Klaus KA, Kumar AA, Fernandez FM, Nair KS. Divergent Skeletal Muscle Metabolomic Signatures of Different Exercise Training Modes Independently Predict Cardiometabolic Risk Factors. Diabetes 2024; 73:23-37. [PMID: 37862464 PMCID: PMC10784655 DOI: 10.2337/db23-0142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 10/09/2023] [Indexed: 10/22/2023]
Abstract
We investigated the link between enhancement of SI (by hyperinsulinemic-euglycemic clamp) and muscle metabolites after 12 weeks of aerobic (high-intensity interval training [HIIT]), resistance training (RT), or combined training (CT) exercise in 52 lean healthy individuals. Muscle RNA sequencing revealed a significant association between SI after both HIIT and RT and the branched-chain amino acid (BCAA) metabolic pathway. Concurrently with increased expression and activity of branched-chain ketoacid dehydrogenase enzyme, many muscle amino metabolites, including BCAAs, glutamate, phenylalanine, aspartate, asparagine, methionine, and γ-aminobutyric acid, increased with HIIT, supporting the substantial impact of HIIT on amino acid metabolism. Short-chain C3 and C5 acylcarnitines were reduced in muscle with all three training modes, but unlike RT, both HIIT and CT increased tricarboxylic acid metabolites and cardiolipins, supporting greater mitochondrial activity with aerobic training. Conversely, RT and CT increased more plasma membrane phospholipids than HIIT, suggesting a resistance exercise effect on cellular membrane protection against environmental damage. Sex and age contributed modestly to the exercise-induced changes in metabolites and their association with cardiometabolic parameters. Integrated transcriptomic and metabolomic analyses suggest various clusters of genes and metabolites are involved in distinct effects of HIIT, RT, and CT. These distinct metabolic signatures of different exercise modes independently link each type of exercise training to improved SI and cardiometabolic risk. ARTICLE HIGHLIGHTS We aimed to understand the link between skeletal muscle metabolites and cardiometabolic health after exercise training. Although aerobic, resistance, and combined exercise training each enhance muscle insulin sensitivity as well as other cardiometabolic parameters, they disparately alter amino and citric acid metabolites as well as the lipidome, linking these metabolomic changes independently to the improvement of cardiometabolic risks with each exercise training mode. These findings reveal an important layer of the unique exercise mode-dependent changes in muscle metabolism, which may eventually lead to more informed exercise prescription for improving SI.
Collapse
Affiliation(s)
- Mark W. Pataky
- Division of Endocrinology and Metabolism, Mayo Clinic, Rochester, MN
| | | | - David A. Gaul
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA
| | - Samuel G. Moore
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA
| | - Surendra Dasari
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN
| | - Matthew M. Robinson
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, OR
| | | | - A. Aneesh Kumar
- Division of Endocrinology and Metabolism, Mayo Clinic, Rochester, MN
| | - Facundo M. Fernandez
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA
| | | |
Collapse
|
22
|
Namwanje M, Mazumdar S, Stayton A, Patel PS, Watkins C, White C, Brown C, Eason JD, Mozhui K, Kuscu C, Pabla N, Stephenson EJ, Bajwa A. Exogenous mitochondrial transfer increases energy expenditure and attenuates adiposity gains in mice with diet-induced obesity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.23.573206. [PMID: 38187751 PMCID: PMC10769436 DOI: 10.1101/2023.12.23.573206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Obesity is associated with chronic multi-system bioenergetic stress that may be improved by increasing the number of healthy mitochondria available across organ systems. However, treatments capable of increasing mitochondrial content are generally limited to endurance exercise training paradigms, which are not always sustainable long-term, let alone feasible for many patients with obesity. Recent studies have shown that local transfer of exogenous mitochondria from healthy donor tissues can improve bioenergetic outcomes and alleviate the effects of tissue injury in recipients with organ specific disease. Thus, the aim of this project was to determine the feasibility of systemic mitochondrial transfer for improving energy balance regulation in the setting of diet-induced obesity. We found that transplantation of mitochondria from lean mice into mice with diet-induced obesity attenuated adiposity gains by increasing energy expenditure and promoting the mobilization and oxidation of lipids. Additionally, mice that received exogenous mitochondria demonstrated improved glucose uptake, greater insulin responsiveness, and complete reversal of hepatic steatosis. These changes were, in part, driven by adaptations occurring in white adipose tissue. Together, these findings are proof-of-principle that mitochondrial transplantation is an effective therapeutic strategy for limiting the deleterious metabolic effects of diet-induced obesity in mice.
Collapse
Affiliation(s)
- Maria Namwanje
- Transplant Research Institute, Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, U.S.A
| | - Soumi Mazumdar
- Transplant Research Institute, Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, U.S.A
| | - Amanda Stayton
- Transplant Research Institute, Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, U.S.A
| | - Prisha S. Patel
- Transplant Research Institute, Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, U.S.A
| | - Christine Watkins
- Transplant Research Institute, Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, U.S.A
| | - Catrina White
- Transplant Research Institute, Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, U.S.A
| | - Chester Brown
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, U.S.A
- Department of Genetics, Genomics, and Informatics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, U.S.A
| | - James D. Eason
- Transplant Research Institute, Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, U.S.A
| | - Khyobeni Mozhui
- Department of Preventive Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, U.S.A
| | - Cem Kuscu
- Transplant Research Institute, Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, U.S.A
| | - Navjot Pabla
- Division of Pharmaceutics and Pharmacology, College of Pharmacy & Comprehensive Cancer Center, The Ohio State University, Columbus, OH, U.S.A
| | - Erin J. Stephenson
- Department of Anatomy, College of Graduate Studies, Midwestern University, Downers Grove, IL, U.S.A
- Physical Therapy Program, College of Health Sciences, Midwestern University, Downers Grove, IL, U.S.A
- Physician Assistant Program, College of Health Sciences, Midwestern University, Downers Grove, IL, U.S.A
- Chicago College of Osteopathic Medicine, Midwestern University, Downers Grove, IL, U.S.A
- College of Dental Medicine Illinois, Midwestern University, Downers Grove, IL, U.S.A
| | - Amandeep Bajwa
- Transplant Research Institute, Department of Surgery, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, U.S.A
- Department of Genetics, Genomics, and Informatics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, U.S.A
- Department of Microbiology, Immunology, and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, U.S.A
| |
Collapse
|
23
|
Tian J, Fan J, Zhang T. Mitochondria as a target for exercise-mitigated type 2 diabetes. J Mol Histol 2023; 54:543-557. [PMID: 37874501 DOI: 10.1007/s10735-023-10158-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 09/17/2023] [Indexed: 10/25/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is one of most common metabolic diseases and continues to be a leading cause of death worldwide. Although great efforts have been made to elucidate the pathogenesis of diabetes, the underlying mechanism still remains unclear. Notably, overwhelming evidence has demonstrated that mitochondria are tightly correlated with the development of T2DM, and the defects of mitochondrial function in peripheral insulin-responsive tissues, such as skeletal muscle, liver and adipose tissue, are crucial drivers of T2DM. Furthermore, exercise training is considered as an effective stimulus for improving insulin sensitivity and hence is regarded as the best strategy to prevent and treat T2DM. Although the precise mechanisms by which exercise alleviates T2DM are not fully understood, mitochondria may be critical for the beneficial effects of exercise.
Collapse
Affiliation(s)
- Jingjing Tian
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai, China
| | - Jingcheng Fan
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai, China
| | - Tan Zhang
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China.
- Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai, China.
| |
Collapse
|
24
|
Schleh MW, Ahn C, Ryan BJ, Chugh OK, Luker AT, Luker KE, Gillen JB, Ludzki AC, Van Pelt DW, Pitchford LM, Zhang T, Rode T, Howton SM, Burant CF, Horowitz JF. Both moderate- and high-intensity exercise training increase intramyocellular lipid droplet abundance and modify myocellular distribution in adults with obesity. Am J Physiol Endocrinol Metab 2023; 325:E466-E479. [PMID: 37729021 PMCID: PMC10864005 DOI: 10.1152/ajpendo.00093.2023] [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: 03/27/2023] [Revised: 09/12/2023] [Accepted: 09/12/2023] [Indexed: 09/22/2023]
Abstract
Exercise training modifies lipid metabolism in skeletal muscle, but the effect of exercise training on intramyocellular lipid droplet (LD) abundance, size, and intracellular distribution in adults with obesity remains elusive. This study compared high-intensity interval training (HIIT) with more conventional moderate-intensity continuous training (MICT) on intramyocellular lipid content, as well as LD characteristics (size and number) and abundance within the intramyofibrillar (IMF) and subsarcolemmal (SS) regions of type I and type II skeletal muscle fibers in adults with obesity. Thirty-six adults with obesity [body mass index (BMI) = 33 ± 3 kg/m2] completed 12 wk (4 days/wk) of either HIIT (10 × 1 min, 90% HRmax + 1-min active recovery; n = 19) or MICT (45-min steady-state exercise, 70% HRmax; n = 17), while on a weight-maintaining diet throughout training. Skeletal muscle biopsies were collected from the vastus lateralis before and after training, and intramyocellular lipid content and intracellular LD distribution were measured by immunofluorescence microscopy. Both MICT and HIIT increased total intramyocellular lipid content by more than 50% (P < 0.01), which was attributed to a greater LD number per µm2 in the IMF region of both type I and type II muscle fibers (P < 0.01). Our findings also suggest that LD lipophagy (autophagy-mediated LD degradation) may be transiently upregulated the day after the last exercise training session (P < 0.02 for both MICT and HIIT). In summary, exercise programs for adults with obesity involving either MICT or HIIT increased skeletal muscle LD abundance via a greater number of LDs in the IMF region of the myocyte, thereby providing more lipid in close proximity to the site of energy production during exercise.NEW & NOTEWORTHY In this study, 12 wk of either moderate-intensity continuous training (MICT) or high-intensity interval training (HIIT) enhanced skeletal muscle lipid abundance by increasing lipid droplet number within the intramyofibrillar (IMF) region of muscle. Because the IMF associates with high energy production during muscle contraction, this adaptation may enhance lipid oxidation during exercise. Despite differences in training intensity and energy expenditure between MICT and HIIT, their effects on muscle lipid abundance and metabolism were remarkably similar.
Collapse
Affiliation(s)
- Michael W Schleh
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Cheehoon Ahn
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Benjamin J Ryan
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Olivia K Chugh
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Austin T Luker
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Kathryn E Luker
- Department of Radiology, Center for Molecular Imaging, University of Michigan, Ann Arbor, Michigan, United States
- Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, United States
| | - Jenna B Gillen
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
- Faculty of Kinesiology and Physical Education, University of Toronto, Toronto, Ontario, Canada
| | - Alison C Ludzki
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Douglas W Van Pelt
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Lisa M Pitchford
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Tao Zhang
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Thomas Rode
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Suzette M Howton
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
| | - Charles F Burant
- Division of Metabolism, Endocrinology, and Diabetes, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, United States
| | - Jeffrey F Horowitz
- Substrate Metabolism Laboratory, School of Kinesiology, University of Michigan, Ann Arbor, Michigan, United States
| |
Collapse
|
25
|
Meyer JN, Pan WK, Ryde IT, Alexander T, Klein-Adams JC, Ndirangu DS, Falvo MJ. Bioenergetic function is decreased in peripheral blood mononuclear cells of veterans with Gulf War Illness. PLoS One 2023; 18:e0287412. [PMID: 37910447 PMCID: PMC10619881 DOI: 10.1371/journal.pone.0287412] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/26/2023] [Indexed: 11/03/2023] Open
Abstract
Gulf War Illness (GWI) is a major health problem for approximately 250,000 Gulf War (GW) veterans, but the etiology of GWI is unclear. We hypothesized that mitochondrial dysfunction is an important contributor to GWI, based on the similarity of some GWI symptoms to those occurring in some mitochondrial diseases; the plausibility that certain pollutants to which GW veterans were exposed affect mitochondria; mitochondrial effects observed in studies in laboratory models of GWI; and previous evidence of mitochondrial outcomes in studies in GW veterans. A primary role of mitochondria is generation of energy via oxidative phosphorylation. However, direct assessment of mitochondrial respiration, reflecting oxidative phosphorylation, has not been carried out in veterans with GWI. In this case-control observational study, we tested multiple measures of mitochondrial function and integrity in a cohort of 114 GW veterans, 80 with and 34 without GWI as assessed by the Kansas definition. In circulating white blood cells, we analyzed multiple measures of mitochondrial respiration and extracellular acidification, a proxy for non-aerobic energy generation; mitochondrial DNA (mtDNA) copy number; mtDNA damage; and nuclear DNA damage. We also collected detailed survey data on demographics; deployment; self-reported exposure to pesticides, pyridostigmine bromide, and chemical and biological warfare agents; and current biometrics, health and activity levels. We observed a 9% increase in mtDNA content in blood in veterans with GWI, but did not detect differences in DNA damage. Basal and ATP-linked oxygen consumption were respectively 42% and 47% higher in veterans without GWI, after adjustment for mtDNA amount. We did not find evidence for a compensatory increase in anaerobic energy generation: extracellular acidification was also lower in GWI (12% lower at baseline). A subset of 27 and 26 veterans returned for second and third visits, allowing us to measure stability of mitochondrial parameters over time. mtDNA CN, mtDNA damage, ATP-linked OCR, and spare respiratory capacity were moderately replicable over time, with intraclass correlation coefficients of 0.43, 0.44, 0.50, and 0.57, respectively. Other measures showed higher visit-to-visit variability. Many measurements showed lower replicability over time among veterans with GWI compared to veterans without GWI. Finally, we found a strong association between recalled exposure to pesticides, pyridostigmine bromide, and chemical and biological warfare agents and GWI (p < 0.01, p < 0.01, and p < 0.0001, respectively). Our results demonstrate decreased mitochondrial respiratory function as well as decreased glycolytic activity, both of which are consistent with decreased energy availability, in peripheral blood mononuclear cells in veterans with GWI.
Collapse
Affiliation(s)
- Joel N. Meyer
- Nicholas School of the Environment, Duke University, Durham, NC, United States of America
| | - William K. Pan
- Nicholas School of the Environment, Duke University, Durham, NC, United States of America
| | - Ian T. Ryde
- Nicholas School of the Environment, Duke University, Durham, NC, United States of America
| | - Thomas Alexander
- Department of Veterans Affairs, War Related Illness and Injury Study Center, East Orange, NJ, United States of America
| | - Jacquelyn C. Klein-Adams
- Department of Veterans Affairs, War Related Illness and Injury Study Center, East Orange, NJ, United States of America
| | - Duncan S. Ndirangu
- Department of Veterans Affairs, War Related Illness and Injury Study Center, East Orange, NJ, United States of America
| | - Michael J. Falvo
- Department of Veterans Affairs, War Related Illness and Injury Study Center, East Orange, NJ, United States of America
- New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, United States of America
| |
Collapse
|
26
|
Whytock KL, Pino MF, Sun Y, Yu G, De Carvalho FG, Yeo RX, Vega RB, Parmar G, Divoux A, Kapoor N, Yi F, Cornnell H, Patten DA, Harper ME, Gardell SJ, Smith SR, Walsh MJ, Sparks LM. Comprehensive interrogation of human skeletal muscle reveals a dissociation between insulin resistance and mitochondrial capacity. Am J Physiol Endocrinol Metab 2023; 325:E291-E302. [PMID: 37584609 PMCID: PMC11901339 DOI: 10.1152/ajpendo.00143.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/10/2023] [Accepted: 08/10/2023] [Indexed: 08/17/2023]
Abstract
Insulin resistance and blunted mitochondrial capacity in skeletal muscle are often synonymous, however, this association remains controversial. The aim of this study was to perform an in-depth multifactorial comparison of skeletal muscle mitochondrial capacity between individuals who were lean and active (Active, n = 9), individuals with obesity (Obese, n = 9), and individuals with obesity, insulin resistance, and type 2 diabetes (T2D, n = 22). Mitochondrial capacity was assessed by ex vivo mitochondrial respiration with fatty-acid and glycolytic-supported protocols adjusted for mitochondrial content (mtDNA and citrate synthase activity). Supercomplex assembly was measured by Blue Native (BN)-PAGE and immunoblot. Tricarboxylic (TCA) cycle intermediates were assessed with targeted metabolomics. Exploratory transcriptomics and DNA methylation analyses were performed to uncover molecular differences affecting mitochondrial function among the three groups. We reveal no discernable differences in skeletal muscle mitochondrial content, mitochondrial capacity, supercomplex assembly, TCA cycle intermediates, and mitochondrial molecular profiles between obese individuals with and without T2D that had comparable levels of confounding factors (body mass index, age, and aerobic capacity). We highlight that lean, active individuals have greater mitochondrial content, mitochondrial capacity, supercomplex assembly, and TCA cycle intermediates. These phenotypical changes are reflected at the level of DNA methylation and gene transcription. The collective observation of comparable muscle mitochondrial capacity in individuals with obesity and T2D (vs. individuals without T2D) underscores a dissociation from skeletal muscle insulin resistance. Clinical trial number: NCT01911104.NEW & NOTEWORTHY Whether impaired mitochondrial capacity contributes to skeletal muscle insulin resistance is debated. Our multifactorial analysis shows no differences in skeletal muscle mitochondrial content, mitochondrial capacity, and mitochondrial molecular profiles between obese individuals with and without T2D that had comparable levels of confounding factors (BMI, age, aerobic capacity). We highlight that lean, active individuals have enhanced skeletal muscle mitochondrial capacity that is also reflected at the level of DNA methylation and gene transcription.
Collapse
Affiliation(s)
- Katie L Whytock
- Translational Research Institute, AdventHealth, Orlando, Florida, United States
| | - Maria F Pino
- Translational Research Institute, AdventHealth, Orlando, Florida, United States
| | - Yifei Sun
- Icahn School of Medicine at Mount Sinai, New York City, New York, United States
| | - GongXin Yu
- Translational Research Institute, AdventHealth, Orlando, Florida, United States
| | | | - Reichelle X Yeo
- Translational Research Institute, AdventHealth, Orlando, Florida, United States
| | - Rick B Vega
- Translational Research Institute, AdventHealth, Orlando, Florida, United States
| | - Gaganvir Parmar
- Department of Biochemistry Microbiology and Immunology, Faculty of Medicine, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Adeline Divoux
- Translational Research Institute, AdventHealth, Orlando, Florida, United States
| | - Nidhi Kapoor
- Translational Research Institute, AdventHealth, Orlando, Florida, United States
| | - Fancaho Yi
- Translational Research Institute, AdventHealth, Orlando, Florida, United States
| | - Heather Cornnell
- Translational Research Institute, AdventHealth, Orlando, Florida, United States
| | - David A Patten
- Department of Biochemistry Microbiology and Immunology, Faculty of Medicine, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Mary-Ellen Harper
- Department of Biochemistry Microbiology and Immunology, Faculty of Medicine, Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - Stephen J Gardell
- Translational Research Institute, AdventHealth, Orlando, Florida, United States
| | - Steven R Smith
- Translational Research Institute, AdventHealth, Orlando, Florida, United States
| | - Martin J Walsh
- Icahn School of Medicine at Mount Sinai, New York City, New York, United States
| | - Lauren M Sparks
- Translational Research Institute, AdventHealth, Orlando, Florida, United States
| |
Collapse
|
27
|
Macedo ACPD, Schaan CW, Bock PM, Pinto MBD, Botton CE, Umpierre D, Schaan BD. Cardiorespiratory fitness in individuals with type 2 diabetes mellitus: a systematic review and meta-analysis. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2023; 67:e230040. [PMID: 37738467 PMCID: PMC10665050 DOI: 10.20945/2359-4292-2023-0040] [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: 01/01/2023] [Accepted: 05/30/2023] [Indexed: 09/24/2023]
Abstract
Objective To conduct a systematic review and meta-analysis assessing the cardiorespiratory fitness (CRF) among individuals with and without type 2 diabetes. Materials and methods The current review was registered in PROSPERO under the number CRD42018082718. MEDLINE, EMBASE, and Cochrane Library databases were searched from inception through February 2022. Eligibility criteria consisted of observational or interventional studies that evaluated CRF through cardiopulmonary exercise testing or six-minute walk test in individuals with type 2 diabetes compared with individuals without type 2 diabetes. For data extraction, we used baseline CRF assessments of randomized clinical trials or follow-up CRF assessments in observational studies. We performed a meta-analysis using maximal oxygen consumption (VO2 max), and distance walked in the 6MWT as primary outcomes. They were extracted and expressed as mean differences (MDs) and 95% CIs between treatment and comparator groups. The meta-analysis was conducted using Review Manager (RevMan) software. Results Out of 8,347 studies retrieved, 77 were included. Compared with individuals without type 2 diabetes, individuals with diabetes achieved a lower VO2 max (-5.84 mL.kg-1.min-1, 95% CI -6.93, -4.76 mL.kg-1.min-1, p = <0.0001; I2 = 91%, p for heterogeneity < 0.0001), and a smaller distance walked in 6MWT (-93.30 meters, 95% CI -141.2, -45.4 meters, p > 0.0001; I2: 94%, p for heterogeneity < 0.0001). Conclusion Type 2 diabetes was associated with lower cardiorespiratory fitness, as observed by lower VO2 max on maximal tests, and smaller distance walked in 6MWT, however the quality of studies was low.
Collapse
Affiliation(s)
- Aline Chagastelles Pinto de Macedo
- Universidade Federal do Rio Grande do Sul, Programa de Pós-graduaçÃo em Ciências Médicas: Endocrinologia, Porto Alegre, RS, Brasil
- Laboratório de Atividade Física, Diabetes e Doença Cardiovascular (LADD), Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| | - Camila Wohlgemuth Schaan
- Laboratório de Atividade Física, Diabetes e Doença Cardiovascular (LADD), Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| | - Patricia Martins Bock
- Laboratório de Atividade Física, Diabetes e Doença Cardiovascular (LADD), Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil,
- Faculdades Integradas de Taquara, Taquara, RS, Brasil
- Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
| | - Mariana Brutto de Pinto
- Laboratório de Atividade Física, Diabetes e Doença Cardiovascular (LADD), Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| | - Cintia Ehlers Botton
- Instituto de AvaliaçÃo de Tecnologia em Saúde (IATS) - CNPq/Brasil, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
- Universidade Federal do Ceará, Instituto de EducaçÃo Física e Esportes, Fortaleza, CE, Brasil
- Programa de Mestrado em Fisioterapia e Funcionalidade, Universidade Federal do Ceará, Fortaleza, CE, Brasil
| | - Daniel Umpierre
- Laboratório de Atividade Física, Diabetes e Doença Cardiovascular (LADD), Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
- Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
- Instituto de AvaliaçÃo de Tecnologia em Saúde (IATS) - CNPq/Brasil, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| | - Beatriz D Schaan
- Universidade Federal do Rio Grande do Sul, Programa de Pós-graduaçÃo em Ciências Médicas: Endocrinologia, Porto Alegre, RS, Brasil
- Laboratório de Atividade Física, Diabetes e Doença Cardiovascular (LADD), Centro de Pesquisa Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
- Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brasil
- Instituto de AvaliaçÃo de Tecnologia em Saúde (IATS) - CNPq/Brasil, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| |
Collapse
|
28
|
Vanni E, Lindner K, Gavin AC, Montessuit C. Differential intracellular management of fatty acids impacts on metabolic stress-stimulated glucose uptake in cardiomyocytes. Sci Rep 2023; 13:14805. [PMID: 37684349 PMCID: PMC10491837 DOI: 10.1038/s41598-023-42072-7] [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: 03/03/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023] Open
Abstract
Stimulation of glucose uptake in response to ischemic metabolic stress is important for cardiomyocyte function and survival. Chronic exposure of cardiomyocytes to fatty acids (FA) impairs the stimulation of glucose uptake, whereas induction of lipid droplets (LD) is associated with preserved glucose uptake. However, the mechanisms by which LD induction prevents glucose uptake impairment remain elusive. We induced LD with either tetradecanoyl phorbol acetate (TPA) or 5-aminoimidazole-4-carboxamide-1-β-D-ribofuranoside (AICAR). Triacylglycerol biosynthesis enzymes were inhibited in cardiomyocytes exposed to FA ± LD inducers, either upstream (glycerol-3-phosphate acyltransferases; GPAT) or downstream (diacylglycerol acyltransferases; DGAT) of the diacylglycerol step. Although both inhibitions reduced LD formation in cardiomyocytes treated with FA and LD inducers, only DGAT inhibition impaired metabolic stress-stimulated glucose uptake. DGAT inhibition in FA plus TPA-treated cardiomyocytes reduced triacylglycerol but not diacylglycerol content, thus increasing the diacylglycerol/triacylglycerol ratio. In cardiomyocytes exposed to FA alone, GPAT inhibition reduced diacylglycerol but not triacylglycerol, thus decreasing the diacylglycerol/triacylglycerol ratio, prevented PKCδ activation and improved metabolic stress-stimulated glucose uptake. Changes in AMP-activated Protein Kinase activity failed to explain variations in metabolic stress-stimulated glucose uptake. Thus, LD formation regulates metabolic stress-stimulated glucose uptake in a manner best reflected by the diacylglycerol/triacylglycerol ratio.
Collapse
Affiliation(s)
- Ettore Vanni
- Department of Pathology and Immunology, University of Geneva School of Medicine, Geneva, Switzerland
| | - Karina Lindner
- Department of Cell Physiology and Metabolism, University of Geneva School of Medicine, Geneva, Switzerland
| | - Anne-Claude Gavin
- Department of Cell Physiology and Metabolism, University of Geneva School of Medicine, Geneva, Switzerland
| | - Christophe Montessuit
- Department of Pathology and Immunology, University of Geneva School of Medicine, Geneva, Switzerland.
| |
Collapse
|
29
|
Al-Rawaf HA, Gabr SA, Iqbal A, Alghadir AH. High-Intensity Interval Training Improves Glycemic Control, Cellular Apoptosis, and Oxidative Stress of Type 2 Diabetic Patients. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1320. [PMID: 37512131 PMCID: PMC10384171 DOI: 10.3390/medicina59071320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/25/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023]
Abstract
Background and Objectives: Physical exercise is an important therapeutic modality for treating and managing diabetes. High-intensity interval training (HIIT) is considered one of the best non-drug strategies for preventing and treating type 2 diabetes mellitus (T2DM) by improving mitochondrial biogenesis and function. This study aimed to determine the effects of 12 weeks of HIIT training on the expression of tumor suppressor protein-p53, mitochondrial cytochrome c oxidase (COX), and oxidative stress in patients with T2DM. Methods: A total of thirty male sedentary patients aged (45-60 years) were diagnosed with established T2DM for more than five years. Twenty healthy volunteers, age- and sex-matched, were included in this study. Both patients and control subjects participated in the HIIT program for 12 weeks. Glycemic control variables including p53 (U/mL), COX (ng/mL), total antioxidant capacity (TAC, nmole/µL), 8-hydroxy-2'-deoxyguanosine (8-OHdG, ng/mL), as well as genomic and mitochondrial DNA content were measured in both the serum and muscle tissues of control and patient groups following exercise training. Results: There were significant improvements in fasting glucose levels. HbA1c (%), HOMA-IR (mUmmol/L2), fasting insulin (µU/mL), and C-peptide (ng/mL) were reported in T2DM and healthy controls. A significant decrease was also observed in p53 protein levels. COX, 8-OhdG, and an increase in the level of TAC were reported in T2DM following 12 weeks of HIIT exercise. Before and after exercise, p53; COX, mt-DNA content, TAC, and 8-OhdG showed an association with diabetic control parameters such as fasting glucose (FG), glycated hemoglobin (HbA1C, %), C-peptide, fasting insulin (FI), and homeostatic model assessment for insulin resistance (HOMA-IR) in patients with T2DM. These findings support the positive impact of HIIT exercise in improving regulation of mitochondrial biogenesis and subsequent control of diabetes through anti-apoptotic and anti-oxidative pathways. Conclusions: A 12-week HIIT program significantly improves diabetes by reducing insulin resistance; regulating mitochondrial biogenesis; and decreasing oxidative stress capacity among patients and healthy controls. Also; p53 protein expression; COX; 8-OhdG; and TAC and mt-DNA content were shown to be associated with T2DM before and after exercise training.
Collapse
Affiliation(s)
- Hadeel A. Al-Rawaf
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia;
| | - Sami A. Gabr
- Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia; (S.A.G.); (A.H.A.)
| | - Amir Iqbal
- Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia; (S.A.G.); (A.H.A.)
| | - Ahmad H. Alghadir
- Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia; (S.A.G.); (A.H.A.)
| |
Collapse
|
30
|
Billon C, Sitaula S, Banerjee S, Welch R, Elgendy B, Hegazy L, Oh TG, Kazantzis M, Chatterjee A, Chrivia J, Hayes ME, Xu W, Hamilton A, Huss JM, Zhang L, Walker JK, Downes M, Evans RM, Burris TP. Synthetic ERRα/β/γ Agonist Induces an ERRα-Dependent Acute Aerobic Exercise Response and Enhances Exercise Capacity. ACS Chem Biol 2023; 18:756-771. [PMID: 36988910 PMCID: PMC11584170 DOI: 10.1021/acschembio.2c00720] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Repetitive physical exercise induces physiological adaptations in skeletal muscle that improves exercise performance and is effective for the prevention and treatment of several diseases. Genetic evidence indicates that the orphan nuclear receptors estrogen receptor-related receptors (ERRs) play an important role in skeletal muscle exercise capacity. Three ERR subtypes exist (ERRα, β, and γ), and although ERRβ/γ agonists have been designed, there have been significant difficulties in designing compounds with ERRα agonist activity. Additionally, there are limited synthetic agonists that can be used to target ERRs in vivo. Here, we report the identification of a synthetic ERR pan agonist, SLU-PP-332, that targets all three ERRs but has the highest potency for ERRα. Additionally, SLU-PP-332 has sufficient pharmacokinetic properties to be used as an in vivo chemical tool. SLU-PP-332 increases mitochondrial function and cellular respiration in a skeletal muscle cell line. When administered to mice, SLU-PP-332 increased the type IIa oxidative skeletal muscle fibers and enhanced exercise endurance. We also observed that SLU-PP-332 induced an ERRα-specific acute aerobic exercise genetic program, and the ERRα activation was critical for enhancing exercise endurance in mice. These data indicate the feasibility of targeting ERRα for the development of compounds that act as exercise mimetics that may be effective in the treatment of numerous metabolic disorders and to improve muscle function in the aging.
Collapse
Affiliation(s)
- Cyrielle Billon
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, Missouri 63110, United States
| | - Sadichha Sitaula
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, Missouri 63110, United States
| | - Subhashis Banerjee
- Department of Pharmacology & Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
| | - Ryan Welch
- Gene Expression Laboratory Salk Institute for Biological Studies, La Jolla, California 92037, United States
| | - Bahaa Elgendy
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, Missouri 63110, United States
| | - Lamees Hegazy
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, Missouri 63110, United States
| | - Tae Gyu Oh
- Gene Expression Laboratory Salk Institute for Biological Studies, La Jolla, California 92037, United States
| | - Melissa Kazantzis
- The Scripps Research Institute Jupiter, Jupiter, Florida 33458, United States
| | - Arindam Chatterjee
- Department of Pharmacology & Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
| | - John Chrivia
- Department of Pharmacology & Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
| | - Matthew E Hayes
- University of Florida Genetics Institute, Gainesville, Florida 32610, United States
| | - Weiyi Xu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, United States
| | - Angelica Hamilton
- Department of Molecular & Cellular Endocrinology, City of Hope, Duarte, California 91010, United States
| | - Janice M Huss
- Department of Molecular & Cellular Endocrinology, City of Hope, Duarte, California 91010, United States
| | - Lilei Zhang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, United States
| | - John K Walker
- Department of Pharmacology & Physiology, Saint Louis University School of Medicine, St. Louis, Missouri 63104, United States
- Department of Chemistry, Saint Louis University, St. Louis, Missouri 63103, United States
| | - Michael Downes
- Gene Expression Laboratory Salk Institute for Biological Studies, La Jolla, California 92037, United States
| | - Ronald M Evans
- Gene Expression Laboratory Salk Institute for Biological Studies, La Jolla, California 92037, United States
- Howard Hughes Medical Institute, Salk Institute for Biological Studies, La Jolla, California 92037, United States
| | - Thomas P Burris
- Center for Clinical Pharmacology, Washington University School of Medicine and St. Louis College of Pharmacy, St. Louis, Missouri 63110, United States
- University of Florida Genetics Institute, Gainesville, Florida 32610, United States
| |
Collapse
|
31
|
Nutrients, Physical Activity, and Mitochondrial Dysfunction in the Setting of Metabolic Syndrome. Nutrients 2023; 15:nu15051217. [PMID: 36904216 PMCID: PMC10004804 DOI: 10.3390/nu15051217] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/04/2023] Open
Abstract
Metabolic syndrome (MetS) is a cluster of metabolic risk factors for diabetes, coronary heart disease, non-alcoholic fatty liver disease, and some tumors. It includes insulin resistance, visceral adiposity, hypertension, and dyslipidemia. MetS is primarily linked to lipotoxicity, with ectopic fat deposition from fat storage exhaustion, more than obesity per se. Excessive intake of long-chain saturated fatty acid and sugar closely relates to lipotoxicity and MetS through several pathways, including toll-like receptor 4 activation, peroxisome proliferator-activated receptor-gamma regulation (PPARγ), sphingolipids remodeling, and protein kinase C activation. These mechanisms prompt mitochondrial dysfunction, which plays a key role in disrupting the metabolism of fatty acids and proteins and in developing insulin resistance. By contrast, the intake of monounsaturated, polyunsaturated, and medium-chain saturated (low-dose) fatty acids, as well as plant-based proteins and whey protein, favors an improvement in sphingolipid composition and metabolic profile. Along with dietary modification, regular exercises including aerobic, resistance, or combined training can target sphingolipid metabolism and improve mitochondrial function and MetS components. This review aimed to summarize the main dietary and biochemical aspects related to the physiopathology of MetS and its implications for mitochondrial machinery while discussing the potential role of diet and exercise in counteracting this complex clustering of metabolic dysfunctions.
Collapse
|
32
|
Role of Skeletal Muscle in the Pathogenesis and Management of Type 2 Diabetes: A Special Focus on Asian Indians. J Indian Inst Sci 2023. [DOI: 10.1007/s41745-022-00349-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
33
|
Buch A, Yeshurun S, Cramer T, Baumann A, Sencelsky Y, Zelber Sagi S, Serebro M, Greenman Y, Mor M, Eldor R. The Effects of Metabolism Tracker Device (Lumen) Usage on Metabolic Control in Adults with Prediabetes: Pilot Clinical Trial. Obes Facts 2023; 16:53-61. [PMID: 36195053 PMCID: PMC9889724 DOI: 10.1159/000527227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 09/05/2022] [Indexed: 02/04/2023] Open
Abstract
INTRODUCTION Prediabetes is a risk factor for type 2 diabetes mellitus (T2DM). However, it may be reversed via lifestyle changes. Lumen is a novel handheld device that measures exhaled CO2 producing results in agreement with those of indirect calorimetry when assessing metabolic fuel usage. The aim of this study was to examine the effects of following Lumen's personalized, measurement-guided lifestyle intervention program on anthropometric and metabolic variables in adults with prediabetes. METHODS A 12-week single-arm intervention study was conducted in 27 participants. Body composition and blood markers were measured at the start and end of the study. Each participant took a daily morning (fasted) measurement and received feedback on their metabolic state (i.e., their degree of fat vs. carbohydrate oxidation). Participants were then provided with personalized daily guidelines for their carbohydrate, fat, and protein consumption, along with recommended lifestyle changes. RESULTS Intention-to-treat analysis revealed a significant decrease in body weight (5.99 kg, p < 0.001), comprising a significant reduction in percentage body fat (2.93%, p < 0.001) and waist circumference (6.23 cm, p < 0.001). Significant reductions were also observed in glycated hemoglobin A1c (0.27%, p < 0.001), triglycerides (0.45 mg/dL, p < 0.001), and systolic blood pressure (0.5 mm Hg, p < 0.05). CONCLUSION In a 12-week pilot study of participants with prediabetes, Lumen usage significantly improved multiple metabolic parameters, demonstrating its potential to deliver better clinical outcomes for patients with T2DM and metabolic syndrome.
Collapse
Affiliation(s)
- Assaf Buch
- Department of Nutritional Sciences, School of Health Sciences, Ariel University, Ariel, Israel
- Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | | | | | | | - Yael Sencelsky
- School of Public Health, University of Haifa, Haifa, Israel
| | | | - Merav Serebro
- Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv Sourasky Medical Center, Tel-Aviv, Israel
- The Sackler Faculty of Medicine Tel-Aviv University, Tel-Aviv, Israel
| | - Yona Greenman
- Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv Sourasky Medical Center, Tel-Aviv, Israel
- The Sackler Faculty of Medicine Tel-Aviv University, Tel-Aviv, Israel
| | | | - Roy Eldor
- Institute of Endocrinology, Metabolism and Hypertension, Tel Aviv Sourasky Medical Center, Tel-Aviv, Israel
- The Sackler Faculty of Medicine Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
34
|
op den Kamp YJ, Gemmink A, de Ligt M, Dautzenberg B, Kornips E, Jorgensen JA, Schaart G, Esterline R, Pava DA, Hoeks J, Schrauwen-Hinderling VB, Kersten S, Havekes B, Koves TR, Muoio DM, Hesselink MK, Oscarsson J, Phielix E, Schrauwen P. Effects of SGLT2 inhibitor dapagliflozin in patients with type 2 diabetes on skeletal muscle cellular metabolism. Mol Metab 2022; 66:101620. [PMID: 36280113 PMCID: PMC9636471 DOI: 10.1016/j.molmet.2022.101620] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/18/2022] [Accepted: 10/19/2022] [Indexed: 11/06/2022] Open
Abstract
OBJECTIVE SGLT2 inhibitors increase urinary glucose excretion and have beneficial effects on cardiovascular and renal outcomes; the underlying mechanism may be metabolic adaptations due to urinary glucose loss. Here, we investigated the cellular and molecular effects of 5 weeks of dapagliflozin treatment on skeletal muscle metabolism in type 2 diabetes patients. METHODS Twenty-six type 2 diabetes mellitus patients were randomized to a 5-week double-blind, cross-over study with 6-8-week wash-out. Skeletal muscle acetylcarnitine levels, intramyocellular lipid (IMCL) content and phosphocreatine (PCr) recovery rate were measured by magnetic resonance spectroscopy (MRS). Ex vivo mitochondrial respiration was measured in skeletal muscle fibers using high resolution respirometry. Intramyocellular lipid droplet and mitochondrial network dynamics were investigated using confocal microscopy. Skeletal muscle levels of acylcarnitines, amino acids and TCA cycle intermediates were measured. Expression of genes involved in fatty acid metabolism were investigated. RESULTS Mitochondrial function, mitochondrial network integrity and citrate synthase and carnitine acetyltransferase activities in skeletal muscle were unaltered after dapagliflozin treatment. Dapagliflozin treatment increased intramyocellular lipid content (0.060 (0.011, 0.110) %, p = 0.019). Myocellular lipid droplets increased in size (0.03 μm2 (0.01-0.06), p < 0.05) and number (0.003 μm-2 (-0.001-0.007), p = 0.09) upon dapagliflozin treatment. CPT1A, CPT1B and malonyl CoA-decarboxylase mRNA expression was increased by dapagliflozin. Fasting acylcarnitine species and C4-OH carnitine levels (0.4704 (0.1246, 0.8162) pmoles∗mg tissue-1, p < 0.001) in skeletal muscle were higher after dapagliflozin treatment, while acetylcarnitine levels were lower (-40.0774 (-64.4766, -15.6782) pmoles∗mg tissue-1, p < 0.001). Fasting levels of several amino acids, succinate, alpha-ketoglutarate and lactate in skeletal muscle were significantly lower after dapagliflozin treatment. CONCLUSION Dapagliflozin treatment for 5 weeks leads to adaptive changes in skeletal muscle substrate metabolism favoring metabolism of fatty acid and ketone bodies and reduced glycolytic flux. The trial is registered with ClinicalTrials.gov, number NCT03338855.
Collapse
Affiliation(s)
| | - Anne Gemmink
- Departments of Nutrition and Movement Sciences, Maastricht, the Netherlands
| | - Marlies de Ligt
- Departments of Nutrition and Movement Sciences, Maastricht, the Netherlands
| | - Bas Dautzenberg
- Departments of Nutrition and Movement Sciences, Maastricht, the Netherlands
| | - Esther Kornips
- Departments of Nutrition and Movement Sciences, Maastricht, the Netherlands
| | | | - Gert Schaart
- Departments of Nutrition and Movement Sciences, Maastricht, the Netherlands
| | | | - Diego A. Pava
- Departments of Nutrition and Movement Sciences, Maastricht, the Netherlands
| | - Joris Hoeks
- Departments of Nutrition and Movement Sciences, Maastricht, the Netherlands
| | - Vera B. Schrauwen-Hinderling
- Departments of Nutrition and Movement Sciences, Maastricht, the Netherlands,Departments of Radiology and Nuclear Medicine, Maastricht, the Netherlands
| | - Sander Kersten
- Division of Human Nutrition and Health, Wageningen University, the Netherlands
| | - Bas Havekes
- Departments of Internal Medicine, Division of Endocrinology, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, 6200 MD Maastricht, the Netherlands
| | - Timothy R. Koves
- Duke Molecular Physiology Institute and the Sarah W. Stedman Nutrition and Metabolism Center, Department of Medicine, Duke University, Durham, NC 27704, USA
| | - Deborah M. Muoio
- Duke Molecular Physiology Institute and the Sarah W. Stedman Nutrition and Metabolism Center, Department of Medicine, Duke University, Durham, NC 27704, USA
| | | | - Jan Oscarsson
- BioPharmaceuticals R&D, Late-Stage Development, Cardiovascular, Renal and Metabolism, AstraZeneca, Gothenburg, Sweden
| | - Esther Phielix
- Departments of Nutrition and Movement Sciences, Maastricht, the Netherlands
| | - Patrick Schrauwen
- Departments of Nutrition and Movement Sciences, Maastricht, the Netherlands,Corresponding author. Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, the Netherlands.
| |
Collapse
|
35
|
Savikj M, Stocks B, Sato S, Caidahl K, Krook A, Deshmukh AS, Zierath JR, Wallberg-Henriksson H. Exercise timing influences multi-tissue metabolome and skeletal muscle proteome profiles in type 2 diabetic patients - A randomized crossover trial. Metabolism 2022; 135:155268. [PMID: 35908579 DOI: 10.1016/j.metabol.2022.155268] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/10/2022] [Accepted: 07/22/2022] [Indexed: 01/06/2023]
Abstract
AIMS/HYPOTHESIS Metabolic effects of exercise may partly depend on the time-of-day when exercise is performed. We tested the hypothesis that exercise timing affects the adaptations in multi-tissue metabolome and skeletal muscle proteome profiles in men with type 2 diabetes. METHODS Men fitting the inclusion (type 2 diabetes, age 45-68 years and body mass index 23-33 kg/m2) and exclusion criteria (insulin treatment, smoking, concurrent systemic disease, and regular exercise training) were included in a randomized crossover trial (n = 15). Participants included in this metabolomics and proteomics analysis fully completed all exercise sessions (n = 8). The trial consisted of two weeks of high-intensity interval training (HIT) (three sessions/week) either in the morning (08:00, n = 5) or afternoon (16:45, n = 3), a two-week wash-out period, and an additional two weeks of HIT at the opposing time. Participants and researchers were not blinded to group allocation. Blood, skeletal muscle and subcutaneous adipose tissue were obtained before the first, and after each training period. Broad-spectrum, untargeted proteomic analysis was performed on skeletal muscle, and metabolomic analysis was performed on all biosamples. Differential content was assessed by linear regression and pathway set enrichment analyses were performed. Coordinated metabolic changes across tissues were identified by Spearman correlation analysis. RESULTS Metabolic and proteomic profiles remained stable after two weeks of HIT, and individual metabolites and proteins were not altered, irrespective of the time of day at which the training was performed. However, coordinated changes in relevant metabolic pathways and protein categories were identified. Morning and afternoon HIT similarly increased plasma diacylglycerols, skeletal muscle acyl-carnitines, and subcutaneous adipose tissue sphingomyelins and lysophospholipids. Acyl-carnitines were central to training-induced metabolic cross-talk across tissues. Plasma carbohydrates, via the penthose phosphate pathway, were increased and skeletal muscle lipids were decreased after morning compared to afternoon HIT. Skeletal muscle lipoproteins were higher, and mitochondrial complex III abundance was lower after morning compared to afternoon HIT. CONCLUSIONS/INTERPRETATION We provide a comprehensive analysis of a multi-tissue metabolomic and skeletal muscle proteomic responses to training at different times of the day in men with type 2 diabetes. Increased circulating lipids and changes in adipose tissue lipid composition were common between morning and afternoon HIT. However, afternoon HIT increased skeletal muscle lipids and mitochondrial content to a greater degree than morning training. Thus, there is a diurnal component in the metabolomic and proteomic response to exercise in men with type 2 diabetes. The clinical relevance of this response warrants further investigation.
Collapse
Affiliation(s)
- Mladen Savikj
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
| | - Ben Stocks
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Shogo Sato
- Center for Epigenetics and Metabolism, INSERM U1233, Department of Biological Chemistry, School of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Kenneth Caidahl
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Krook
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Atul S Deshmukh
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; The Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Juleen R Zierath
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden; Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
36
|
ANT1 overexpression models: Some similarities with facioscapulohumeral muscular dystrophy. Redox Biol 2022; 56:102450. [PMID: 36030628 PMCID: PMC9434167 DOI: 10.1016/j.redox.2022.102450] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/04/2022] [Accepted: 08/17/2022] [Indexed: 11/20/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant disorder characterized by progressive muscle weakness. Adenine nucleotide translocator 1 (ANT1), the only 4q35 gene involved in mitochondrial function, is strongly expressed in FSHD skeletal muscle biopsies. However, its role in FSHD is unclear. In this study, we evaluated ANT1 overexpression effects in primary myoblasts from healthy controls and during Xenopus laevis organogenesis. We also compared ANT1 overexpression effects with the phenotype of FSHD muscle cells and biopsies. Here, we report that the ANT1 overexpression-induced phenotype presents some similarities with FSHD muscle cells and biopsies. ANT1-overexpressing muscle cells showed disorganized morphology, altered cytoskeletal arrangement, enhanced mitochondrial respiration/glycolysis, ROS production, oxidative stress, mitochondrial fragmentation and ultrastructure alteration, as observed in FSHD muscle cells. ANT1 overexpression in Xenopus laevis embryos affected skeletal muscle development, impaired skeletal muscle, altered mitochondrial ultrastructure and led to oxidative stress as observed in FSHD muscle biopsies. Moreover, ANT1 overexpression in X. laevis embryos affected heart structure and mitochondrial ultrastructure leading to cardiac arrhythmia, as described in some patients with FSHD. Overall our data suggest that ANT1 could contribute to mitochondria dysfunction and oxidative stress in FSHD muscle cells by modifying their bioenergetic profile associated with ROS production. Such interplay between energy metabolism and ROS production in FSHD will be of significant interest for future prospects.
Collapse
|
37
|
Macrophage Polarization Mediated by Mitochondrial Dysfunction Induces Adipose Tissue Inflammation in Obesity. Int J Mol Sci 2022; 23:ijms23169252. [PMID: 36012516 PMCID: PMC9409464 DOI: 10.3390/ijms23169252] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 12/06/2022] Open
Abstract
Obesity is one of the prominent global health issues, contributing to the growing prevalence of insulin resistance and type 2 diabetes. Chronic inflammation in adipose tissue is considered as a key risk factor for the development of insulin resistance and type 2 diabetes in obese individuals. Macrophages are the most abundant immune cells in adipose tissue and play an important role in adipose tissue inflammation. Mitochondria are critical for regulating macrophage polarization, differentiation, and survival. Changes to mitochondrial metabolism and physiology induced by extracellular signals may underlie the corresponding state of macrophage activation. Macrophage mitochondrial dysfunction is a key mediator of obesity-induced macrophage inflammatory response and subsequent systemic insulin resistance. Mitochondrial dysfunction drives the activation of the NLRP3 inflammasome, which induces the release of IL-1β. IL-1β leads to decreased insulin sensitivity of insulin target cells via paracrine signaling or infiltration into the systemic circulation. In this review, we discuss the new findings on how obesity induces macrophage mitochondrial dysfunction and how mitochondrial dysfunction induces NLRP3 inflammasome activation. We also summarize therapeutic approaches targeting mitochondria for the treatment of diabetes.
Collapse
|
38
|
Zhou Y, Wu W, Zou Y, Huang W, Lin S, Ye J, Lan Y. Benefits of different combinations of aerobic and resistance exercise for improving plasma glucose and lipid metabolism and sleep quality among elderly patients with metabolic syndrome: a randomized controlled trial. Endocr J 2022; 69:819-830. [PMID: 35197411 DOI: 10.1507/endocrj.ej21-0589] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Exercise has beneficial effects on metabolic syndrome (MS). However, the exercise prescriptions that best support plasma glucose and lipid control remain unknown. We evaluated the effects of different combinations of aerobic and resistance training programs on plasma glucose and lipid metabolism and sleep quality in elderly MS patients. Eighty-five elderly MS patients were randomly assigned to five groups: aerobic training (AT), resistance training (RT), high aerobic with low resistance training (HALRT), high resistance with low aerobic training (HRLAT), or control. The exercise groups performed supervised moderate-intensity exercise during three 50-min sessions per week for 12 weeks. Body mass index (BMI), waist circumference (WC), systolic blood pressure (SBP), diastolic blood pressure (DBP), handgrip strength (HGS), fasting plasma glucose (FPG), 2-hour postprandial blood glucose (2hPG), total cholesterol (TC), triglyceride (TG), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C) levels and sleep quality were evaluated at baseline and after 12 weeks. All intervention groups showed significant improvements in SBP, HGS, FPG, 2hPG, and Pittsburgh Sleep Quality Index (PSQI) scores compared to baseline (all p < 0.05), while DBP, TC, TG, and LDL-C levels were significantly improved only in the HRLAT and HALRT groups (p < 0.05). The HALRT group showed the largest improvements in WC, SBP, DBP, HGS, FPG, 2hPG, and PSQI score (p < 0.001). The largest improvements in BMI, TC, and LDL-C were observed in the HRLAT group (p < 0.001). The combined exercise prescriptions were more effective than aerobic or resistance training alone at improving plasma glucose and lipid metabolism and sleep quality in elderly MS patients.
Collapse
Affiliation(s)
- Ye Zhou
- School of Nursing, Guangdong Pharmaceutical University, Guangzhou, 510000, China
| | - Wanling Wu
- Hui Ya Hospital of The First Affiliated Hospital, Sun Yat-sen University, Huizhou, 516003, China
| | - Youqing Zou
- The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, 519099, China
| | - Wentao Huang
- School of Nursing, Guangdong Pharmaceutical University, Guangzhou, 510000, China
| | - Shanshan Lin
- Faculty of Health, University of Technology Sydney, Sydney, 201101, Australia
| | - Jinsong Ye
- Taikang Yueyuan Nursing Home, Guangzhou, 510000, China
| | - Yutao Lan
- School of Nursing, Guangdong Pharmaceutical University, Guangzhou, 510000, China
| |
Collapse
|
39
|
Stocks B, Zierath JR. Post-translational Modifications: The Signals at the Intersection of Exercise, Glucose Uptake, and Insulin Sensitivity. Endocr Rev 2022; 43:654-677. [PMID: 34730177 PMCID: PMC9277643 DOI: 10.1210/endrev/bnab038] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Indexed: 11/19/2022]
Abstract
Diabetes is a global epidemic, of which type 2 diabetes makes up the majority of cases. Nonetheless, for some individuals, type 2 diabetes is eminently preventable and treatable via lifestyle interventions. Glucose uptake into skeletal muscle increases during and in recovery from exercise, with exercise effective at controlling glucose homeostasis in individuals with type 2 diabetes. Furthermore, acute and chronic exercise sensitizes skeletal muscle to insulin. A complex network of signals converge and interact to regulate glucose metabolism and insulin sensitivity in response to exercise. Numerous forms of post-translational modifications (eg, phosphorylation, ubiquitination, acetylation, ribosylation, and more) are regulated by exercise. Here we review the current state of the art of the role of post-translational modifications in transducing exercise-induced signals to modulate glucose uptake and insulin sensitivity within skeletal muscle. Furthermore, we consider emerging evidence for noncanonical signaling in the control of glucose homeostasis and the potential for regulation by exercise. While exercise is clearly an effective intervention to reduce glycemia and improve insulin sensitivity, the insulin- and exercise-sensitive signaling networks orchestrating this biology are not fully clarified. Elucidation of the complex proteome-wide interactions between post-translational modifications and the associated functional implications will identify mechanisms by which exercise regulates glucose homeostasis and insulin sensitivity. In doing so, this knowledge should illuminate novel therapeutic targets to enhance insulin sensitivity for the clinical management of type 2 diabetes.
Collapse
Affiliation(s)
- Ben Stocks
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Juleen R Zierath
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark.,Departments of Molecular Medicine and Surgery and Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
40
|
Yeo RX, Dijkstra PJ, De Carvalho FG, Yi F, Pino MF, Smith SR, Sparks LM. Aerobic training increases mitochondrial respiratory capacity in human skeletal muscle stem cells from sedentary individuals. Am J Physiol Cell Physiol 2022; 323:C606-C616. [PMID: 35785986 DOI: 10.1152/ajpcell.00146.2022] [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: 11/22/2022]
Abstract
The impact of aerobic training on human skeletal muscle cell (HSkMC) mitochondrial metabolism is a significant research gap, critical to understanding the mechanisms by which exercise augments skeletal muscle metabolism. We therefore assessed mitochondrial content and capacity in fully differentiated CD56+ HSkMCs from lean active (LA) and sedentary individuals with obesity (OS) at baseline, as well as lean/overweight sedentary individuals (LOS) at baseline and following an 18-day aerobic training intervention. Participants had in vivo skeletal muscle PCr recovery rate by 31P-MRS (mitochondrial oxidative kinetics) and cardiorespiratory fitness (VO2max) assessed at baseline. Biopsies of the vastus lateralis were performed for the isolation of skeletal muscle stem cells. LOS individuals repeated all assessments post-training. HSkMCs were evaluated for mitochondrial respiratory capacity by high resolution respirometry. Data were normalized to two indices of mitochondrial content (CS activity and OXPHOS protein expression) and a marker of total cell count (quantity of DNA).LA individuals had significantly higher VO2max than OS and LOS-Pre training; however, no differences were observed in skeletal muscle mitochondrial capacity, nor in carbohydrate- or fatty acid-supported HSkMC respiratory capacity. Aerobic training robustly increased in vivo skeletal muscle mitochondrial capacity of LOS individuals, as well as carbohydrate-supported HSkMC respiratory capacity. Indices of mitochondrial content and total cell count were similar among the groups and did not change with aerobic training.Our findings demonstrate that bioenergetic changes induced with aerobic training in skeletal muscle in vivo are retained in HSkMCs in vitro without impacting mitochondrial content, suggesting that training improves intrinsic skeletal muscle mitochondrial capacity.
Collapse
Affiliation(s)
- Reichelle X Yeo
- AdventHealth Translational Research Institute, Orlando, FL, United States
| | - Pieter J Dijkstra
- AdventHealth Translational Research Institute, Orlando, FL, United States
| | | | - Fanchao Yi
- AdventHealth Translational Research Institute, Orlando, FL, United States
| | - Maria F Pino
- AdventHealth Translational Research Institute, Orlando, FL, United States
| | - Steven R Smith
- AdventHealth Translational Research Institute, Orlando, FL, United States
| | - Lauren M Sparks
- AdventHealth Translational Research Institute, Orlando, FL, United States
| |
Collapse
|
41
|
Vanweert F, Neinast M, Tapia EE, van de Weijer T, Hoeks J, Schrauwen-Hinderling VB, Blair MC, Bornstein MR, Hesselink MKC, Schrauwen P, Arany Z, Phielix E. A randomized placebo-controlled clinical trial for pharmacological activation of BCAA catabolism in patients with type 2 diabetes. Nat Commun 2022; 13:3508. [PMID: 35717342 PMCID: PMC9206682 DOI: 10.1038/s41467-022-31249-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 06/06/2022] [Indexed: 01/07/2023] Open
Abstract
Elevations in plasma branched-chain amino acid (BCAA) levels associate with insulin resistance and type 2 diabetes (T2D). Pre-clinical models suggest that lowering BCAA levels improve glucose tolerance, but data in humans are lacking. Here, we used sodium phenylbutyrate (NaPB), an accelerator of BCAA catabolism, as tool to lower plasma BCAA levels in patients with T2D, and evaluate its effect on metabolic health. This trial (NetherlandsTrialRegister: NTR7426) had a randomized, placebo-controlled, double-blind cross-over design and was performed in the Maastricht University Medical Center (MUMC+), the Netherlands, between February 2019 and February 2020. Patients were eligible for the trial if they were 40-75years, BMI of 25-38 kg/m², relatively well-controlled T2D (HbA1C < 8.5%) and treated with oral glucose-lowering medication. Eighteen participants were randomly assigned to receive either NaPB 4.8 g/m²/day and placebo for 2 weeks via controlled randomization and sixteen participants completed the study. The primary outcome was peripheral insulin sensitivity. Secondary outcomes were ex vivo muscle mitochondrial oxidative capacity, substrate oxidation and ectopic fat accumulation. Fasting blood samples were collected to determine levels of BCAA, their catabolic intermediates, insulin, triglycerides, free fatty acids (FFA) and glucose. NaPB led to a robust 27% improvement in peripheral insulin sensitivity compared to placebo (ΔRd:13.2 ± 1.8 vs. 9.6 ± 1.8 µmol/kg/min, p = 0.02). This was paralleled by an improvement in pyruvate-driven muscle mitochondrial oxidative capacity and whole-body insulin-stimulated carbohydrate oxidation, and a reduction in plasma BCAA and glucose levels. No effects were observed on levels of insulin, triglycerides and FFA, neither did fat accumulation in muscle and liver change. No adverse events were reported. These data establish the proof-of-concept in humans that modulating the BCAA oxidative pathway may represent a potential treatment strategy for patients with T2D.
Collapse
Affiliation(s)
- Froukje Vanweert
- grid.5012.60000 0001 0481 6099Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, 6229 ER The Netherlands
| | - Michael Neinast
- grid.25879.310000 0004 1936 8972Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, PA 19104 USA
| | - Edmundo Erazo Tapia
- grid.5012.60000 0001 0481 6099Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, 6229 ER The Netherlands
| | - Tineke van de Weijer
- grid.5012.60000 0001 0481 6099Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, 6229 ER The Netherlands ,grid.412966.e0000 0004 0480 1382Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, 6229 ER The Netherlands
| | - Joris Hoeks
- grid.5012.60000 0001 0481 6099Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, 6229 ER The Netherlands
| | - Vera B. Schrauwen-Hinderling
- grid.5012.60000 0001 0481 6099Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, 6229 ER The Netherlands ,grid.412966.e0000 0004 0480 1382Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, 6229 ER The Netherlands
| | - Megan C. Blair
- grid.25879.310000 0004 1936 8972Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, PA 19104 USA
| | - Marc R. Bornstein
- grid.25879.310000 0004 1936 8972Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, PA 19104 USA
| | - Matthijs K. C. Hesselink
- grid.5012.60000 0001 0481 6099Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, 6229 ER The Netherlands
| | - Patrick Schrauwen
- grid.5012.60000 0001 0481 6099Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, 6229 ER The Netherlands
| | - Zoltan Arany
- grid.25879.310000 0004 1936 8972Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, PA 19104 USA
| | - Esther Phielix
- grid.5012.60000 0001 0481 6099Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University, Maastricht, 6229 ER The Netherlands
| |
Collapse
|
42
|
Pirani H, Roustaie M, Ravasi AA, Lamir AR. Effects of 8-week high-intensity interval training and continuous aerobic training on asprosin secretion and fibrillin-1 gene expression levels in diabetic male rats. Int J Diabetes Dev Ctries 2022. [DOI: 10.1007/s13410-022-01056-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
43
|
Gildea N, McDermott A, Rocha J, Crognale D, Nevin A, O'Shea D, Green S, Egaña M. Low-volume HIIT and MICT speed V̇O 2 kinetics during high-intensity "work-to-work" cycling with a similar time-course in type 2 diabetes. J Appl Physiol (1985) 2022; 133:273-287. [PMID: 35678744 DOI: 10.1152/japplphysiol.00148.2022] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We assessed the rates of adjustment in oxygen uptake (V̇O2) and muscle deoxygenation (i.e., deoxygenated haemoglobin and myoglobin, [HHb+Mb]) during the on-transition to high-intensity cycling initiated from an elevated baseline (work-to-work) before training and at weeks 3, 6, 9 and 12 of low-volume high-intensity interval training (HIIT) and moderate-intensity continuous training (MICT) in type 2 diabetes (T2D). Participants were randomly assigned to MICT (n=11, 50 min of moderate-intensity cycling), HIIT (n =8, 10x1 min of high-intensity cycling separated by 1-min of light cycling) or non-exercising control (n=9) groups. Exercising groups trained 3 times per week. Participants completed two work-to-work transitions at each time point consisting of sequential step increments to moderate- and high-intensity work-rates. [HHb+Mb] kinetics were measured by near-infrared spectroscopy at the vastus lateralis muscle. The pretraining time constant of the primary phase of V̇O2 (V̇O2τp) and the amplitude of the V̇O2 slow component (V̇O2As) of the high-intensity w-to-w bout decreased (P<0.05) by a similar magnitude at wk 3 of training in both MICT (from, 56±9 to 43±6s, and from 0.17±0.07 to 0.09±0.05 L.min-1, respectively) and HIIT (from, 56±8 to 42±6s, and from 0.18±0.05 to 0.09±0.08 L.min-1, respectively) with no further changes thereafter. No changes were reported in controls. The parameter estimates of Δ[HHb+Mb] remained unchanged in all groups. MICT and HIIT elicited comparable improvements in V̇O2 kinetics without changes in muscle deoxygenation kinetics during high-intensity exercise initiated from an elevated baseline in T2D despite training volume and time commitment being ~50% lower in the HIIT group.
Collapse
Affiliation(s)
- Norita Gildea
- Department of Physiology, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Adam McDermott
- Department of Physiology, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Joel Rocha
- Division of Sport and Exercise Sciences, School of Applied Sciences, Abertay University, Dundee, United Kingdom
| | - Domenico Crognale
- Institute for Sport and Health, School of Public Health, Physiotherapy and Sports Science, University College Dublin, Ireland
| | - Aaron Nevin
- Department of Physiology, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| | - Donal O'Shea
- Department of Endocrinology, St. Columcille's Hospital, Dublin, Ireland.,Department of Endocrinology and Diabetes Mellitus, St. Vincent's University Hospital, Dublin, Ireland
| | - Simon Green
- Schools of Health Sciences and Medicine, Western Sydney University, Sydney, Australia
| | - Mikel Egaña
- Department of Physiology, School of Medicine, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
44
|
Singh A, D'Amico D, Andreux PA, Fouassier AM, Blanco-Bose W, Evans M, Aebischer P, Auwerx J, Rinsch C. Urolithin A improves muscle strength, exercise performance, and biomarkers of mitochondrial health in a randomized trial in middle-aged adults. Cell Rep Med 2022; 3:100633. [PMID: 35584623 PMCID: PMC9133463 DOI: 10.1016/j.xcrm.2022.100633] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 02/24/2022] [Accepted: 04/21/2022] [Indexed: 12/14/2022]
Abstract
Targeting mitophagy to activate the recycling of faulty mitochondria during aging is a strategy to mitigate muscle decline. We present results from a randomized, placebo-controlled trial in middle-aged adults where we administer a postbiotic compound Urolithin A (Mitopure), a known mitophagy activator, at two doses for 4 months (NCT03464500). The data show significant improvements in muscle strength (∼12%) with intake of Urolithin A. We observe clinically meaningful improvements with Urolithin A on aerobic endurance (peak oxygen oxygen consumption [VO2]) and physical performance (6 min walk test) but do not notice a significant improvement on peak power output (primary endpoint). Levels of plasma acylcarnitines and C-reactive proteins are significantly lower with Urolithin A, indicating higher mitochondrial efficiency and reduced inflammation. We also examine expression of proteins linked to mitophagy and mitochondrial metabolism in skeletal muscle and find a significant increase with Urolithin A administration. This study highlights the benefit of Urolithin A to improve muscle performance. Oral supplementation with Urolithin A increases muscle strength High dose of Urolithin A positively impacts exercise-performance measures An increase in mitophagy proteins in human skeletal muscle observed in parallel Supplementation is safe and increases circulating levels of Urolithin A
Collapse
Affiliation(s)
- Anurag Singh
- Amazentis SA, EPFL Innovation Park, Bâtiment C, 1015 Lausanne, Switzerland.
| | - Davide D'Amico
- Amazentis SA, EPFL Innovation Park, Bâtiment C, 1015 Lausanne, Switzerland
| | - Pénélope A Andreux
- Amazentis SA, EPFL Innovation Park, Bâtiment C, 1015 Lausanne, Switzerland
| | | | | | - Mal Evans
- KGK Science, 255 Queens Avenue #1440, London, ON N6A 5R8, Canada
| | - Patrick Aebischer
- Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Chris Rinsch
- Amazentis SA, EPFL Innovation Park, Bâtiment C, 1015 Lausanne, Switzerland
| |
Collapse
|
45
|
Georgiev A, Granata C, Roden M. The role of mitochondria in the pathophysiology and treatment of common metabolic diseases in humans. Am J Physiol Cell Physiol 2022; 322:C1248-C1259. [PMID: 35508191 DOI: 10.1152/ajpcell.00035.2022] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Common metabolic diseases such as obesity, type 2 diabetes mellitus and non-alcoholic fatty liver disease significantly contribute to morbidity and mortality worldwide. They frequently associate with insulin resistance and altered mitochondrial functionality. Insulin-responsive tissues can show changes in mitochondrial features such as oxidative capacity, mitochondrial content and turnover, which do not necessarily reflect abnormalities but rather adaption to a certain metabolic condition. Lifestyle modifications and classic or novel drugs can modify these alterations and help treating these metabolic diseases. This review addresses the role of mitochondria in human metabolic diseases and discusses potential future research directions.
Collapse
Affiliation(s)
- Asen Georgiev
- Institute for Clinical Diabetology, German, Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany
| | - Cesare Granata
- Institute for Clinical Diabetology, German, Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany.,Department of Diabetes, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Institute for Health and Sport (iHeS), Victoria University, Melbourne, VIC, Australia
| | - Michael Roden
- Institute for Clinical Diabetology, German, Diabetes Center, Leibniz Center for Diabetes Research at Heinrich-Heine-University, Düsseldorf, Düsseldorf, Germany.,German Center for Diabetes Research, Partner Düsseldorf, München-Neuherberg, Germany.,Department of Endocrinology and Diabetology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
46
|
Brugnara L, García AI, Murillo S, Ribalta J, Fernandez G, Marquez S, Rodriguez MA, Vinaixa M, Amigó N, Correig X, Kalko S, Pomes J, Novials A. Muscular carnosine is a marker for cardiorespiratory fitness and cardiometabolic risk factors in men with type 1 diabetes. Eur J Appl Physiol 2022; 122:1429-1440. [PMID: 35298695 DOI: 10.1007/s00421-022-04929-z] [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: 04/13/2021] [Accepted: 03/04/2022] [Indexed: 12/01/2022]
Abstract
PURPOSE Muscle is an essential organ for glucose metabolism and can be influenced by metabolic disorders and physical activity. Elevated muscle carnosine levels have been associated with insulin resistance and cardiometabolic risk factors. Little is known about muscle carnosine in type 1 diabetes (T1D) and how it is influenced by physical activity. The aim of this study was to characterize muscle carnosine in vivo by proton magnetic resonance spectroscopy (1H MRS) and evaluate the relationship with physical activity, clinical characteristics and lipoprotein subfractions. METHODS 16 men with T1D (10 athletes/6 sedentary) and 14 controls without diabetes (9/5) were included. Body composition by DXA, cardiorespiratory capacity (VO2peak) and serum lipoprotein profile by proton nuclear magnetic resonance (1H NMR) were obtained. Muscle carnosine scaled to water (carnosineW) and to creatine (carnosineCR), creatine and intramyocellular lipids (IMCL) were quantified in vivo using 1H MRS in a 3T MR scanner in soleus muscle. RESULTS Subjects with T1D presented higher carnosine CR levels compared to controls. T1D patients with a lower VO2peak presented higher carnosineCR levels compared to sedentary controls, but both T1D and control groups presented similar levels of carnosineCR at high VO2peak levels. CarnosineW followed the same trend. Integrated correlation networks in T1D demonstrated that carnosineW and carnosineCR were associated with cardiometabolic risk factors including total and abdominal fat, pro-atherogenic lipoproteins (very low-density lipoprotein subfractions), low VO2peak, and IMCL. CONCLUSIONS Elevated muscle carnosine levels in persons with T1D and their effect on atherogenic lipoproteins can be modulated by physical activity.
Collapse
Affiliation(s)
- Laura Brugnara
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Hospital Clínic de Barcelona, Carrer del Rosselló, 149, 08036, Barcelona, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Ana Isabel García
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Hospital Clínic de Barcelona, Carrer del Rosselló, 149, 08036, Barcelona, Spain.,Department of Radiology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Serafín Murillo
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Hospital Clínic de Barcelona, Carrer del Rosselló, 149, 08036, Barcelona, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Josep Ribalta
- Departament de Medicina i Cirugia, Universitat Rovira i Virgili/Unitat de Recerca en Lípids i Arteriosclerosi, IISRV, Reus, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Guerau Fernandez
- Bioinformatics Unit, Genetics and Molecular Medicine Service, Hospital Sant Joan de Déu, Esplugues de Llobregat, Spain
| | - Susanna Marquez
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Maria Vinaixa
- Metabolomics Platform, Universitat Rovira i Virgili, IISRV, Reus, Spain
| | - Núria Amigó
- Metabolomics Platform, Universitat Rovira i Virgili, IISRV, Reus, Spain.,Biosfer Teslab, Reus, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Xavier Correig
- Metabolomics Platform, Universitat Rovira i Virgili, IISRV, Reus, Spain.,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain
| | - Susana Kalko
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Hospital Clínic de Barcelona, Carrer del Rosselló, 149, 08036, Barcelona, Spain.,Bioinformatics Core Facility (IDIBAPS), Barcelona, Spain
| | - Jaume Pomes
- Department of Radiology, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Anna Novials
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Hospital Clínic de Barcelona, Carrer del Rosselló, 149, 08036, Barcelona, Spain. .,Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders (CIBERDEM), Madrid, Spain.
| |
Collapse
|
47
|
Mthembu SXH, Mazibuko-Mbeje SE, Ziqubu K, Nyawo TA, Obonye N, Nyambuya TM, Nkambule BB, Silvestri S, Tiano L, Muller CJF, Dludla PV. Impact of physical exercise and caloric restriction in patients with type 2 diabetes: Skeletal muscle insulin resistance and mitochondrial dysfunction as ideal therapeutic targets. Life Sci 2022; 297:120467. [PMID: 35271881 DOI: 10.1016/j.lfs.2022.120467] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/04/2022] [Accepted: 03/04/2022] [Indexed: 01/01/2023]
Abstract
Skeletal muscle insulin resistance and mitochondrial dysfunction are some of the major pathological defects implicated in the development of type 2 diabetes (T2D). Therefore, it has become necessary to understand how common interventions such as physical exercise and caloric restriction affect metabolic function, including physiological processes that implicate skeletal muscle dysfunction within a state of T2D. This review critically discusses evidence on the impact of physical exercise and caloric restriction on markers of insulin resistance and mitochondrial dysfunction within the skeletal muscle of patients with T2D or related metabolic complications. Importantly, relevant information from clinical studies was acquired through a systematic approach targeting major electronic databases and search engines such as PubMed, Google Scholar, and Cochrane library. The reported evidence suggests that interventions like physical exercise and caloric restriction, within a duration of approximately 2 to 4 months, can improve insulin sensitivity, in part by targeting the phosphoinositide 3-kinases/protein kinase B pathway in patients with T2D. Furthermore, both physical exercise and caloric restriction can effectively modulate markers related to improved mitochondrial function and dynamics. This was consistent with an improved modulation of mitochondrial oxidative capacity and reduced production of reactive oxygen species in patients with T2D or related metabolic complications. However, such conclusions are based on limited evidence, additional clinical trials are required to better understand these interventions on pathological mechanisms of T2D and related abnormalities.
Collapse
Affiliation(s)
- Sinenhlanhla X H Mthembu
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa; Department of Biochemistry, North-West University, Mafikeng Campus, Mmabatho 2735, South Africa
| | | | - Khanyisani Ziqubu
- Department of Biochemistry, North-West University, Mafikeng Campus, Mmabatho 2735, South Africa
| | - Thembeka A Nyawo
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa; Centre for Cardiometabolic Research Africa (CARMA), Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa
| | - Nnini Obonye
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa; Centre for Cardiometabolic Research Africa (CARMA), Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa
| | - Tawanda M Nyambuya
- Department of Health Sciences, Faculty of Health and Applied Sciences, Namibia University of Science and Technology, Windhoek 9000, Namibia
| | - Bongani B Nkambule
- School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu Natal, Durban 4000, South Africa
| | - Sonia Silvestri
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy
| | - Luca Tiano
- Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy
| | - Christo J F Muller
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa; Centre for Cardiometabolic Research Africa (CARMA), Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg 7505, South Africa; Department of Biochemistry and Microbiology, University of Zululand, KwaDlangezwa 3880, South Africa
| | - Phiwayinkosi V Dludla
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa.
| |
Collapse
|
48
|
Scalzo RL, Schauer IE, Rafferty D, Knaub LA, Kvaratskhelia N, Johnson TK, Pott GB, Abushamat LA, Whipple MO, Huebschmann AG, Cree-Green M, Reusch JEB, Regensteiner JG. Single-leg exercise training augments in vivo skeletal muscle oxidative flux and vascular content and function in adults with type 2 diabetes. J Physiol 2022; 600:963-978. [PMID: 33569797 PMCID: PMC9006339 DOI: 10.1113/jp280603] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 01/28/2021] [Indexed: 12/14/2022] Open
Abstract
KEY POINTS People with type 2 diabetes (T2D) have impaired skeletal muscle oxidative flux due to limited oxygen delivery. In the current study, this impairment in oxidative flux in people with T2D was abrogated with a single-leg exercise training protocol. Additionally, single-leg exercise training increased skeletal muscle CD31 content, calf blood flow and state 4 mitochondrial respiration in all participants. ABSTRACT Cardiorespiratory fitness is impaired in type 2 diabetes (T2D), conferring significant cardiovascular risk in this population; interventions are needed. Previously, we reported that a T2D-associated decrement in skeletal muscle oxidative flux is ameliorated with acute use of supplemental oxygen, suggesting that skeletal muscle oxygenation is rate-limiting to in vivo mitochondrial oxidative flux during exercise in T2D. We hypothesized that single-leg exercise training (SLET) would improve the T2D-specific impairment in in vivo mitochondrial oxidative flux during exercise. Adults with (n = 19) and without T2D (n = 22) with similar body mass indexes and levels of physical activity participated in two weeks of SLET. Following SLET, in vivo oxidative flux measured by 31 P-MRS increased in participants with T2D, but not people without T2D, measured by the increase in initial phosphocreatine synthesis (P = 0.0455 for the group × exercise interaction) and maximum rate of oxidative ATP synthesis (P = 0.0286 for the interaction). Additionally, oxidative phosphorylation increased in all participants with SLET (P = 0.0209). After SLET, there was no effect of supplemental oxygen on any of the in vivo oxidative flux measurements in either group (P > 0.02), consistent with resolution of the T2D-associated oxygen limitation previously observed at baseline in subjects with T2D. State 4 mitochondrial respiration also improved in muscle fibres ex vivo. Skeletal muscle vasculature content and calf blood flow increased in all participants with SLET (P < 0.0040); oxygen extraction in the calf increased only in T2D (P = 0.0461). SLET resolves the T2D-associated impairment of skeletal muscle in vivo mitochondrial oxidative flux potentially through improved effective blood flow/oxygen delivery.
Collapse
Affiliation(s)
- Rebecca L Scalzo
- Division of Endocrinology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Division of Center for Women's Health Research, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
- Rocky Mountain Regional Veterans Administration Medical Center, Aurora, Colorado, USA
| | - Irene E Schauer
- Division of Endocrinology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Rocky Mountain Regional Veterans Administration Medical Center, Aurora, Colorado, USA
| | - Deirdre Rafferty
- Division of General Internal Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Leslie A Knaub
- Division of Endocrinology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Rocky Mountain Regional Veterans Administration Medical Center, Aurora, Colorado, USA
| | - Nina Kvaratskhelia
- Division of Endocrinology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Taro Kaelix Johnson
- Division of Endocrinology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Gregory B Pott
- Division of Endocrinology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Rocky Mountain Regional Veterans Administration Medical Center, Aurora, Colorado, USA
| | - Layla A Abushamat
- Division of Endocrinology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Rocky Mountain Regional Veterans Administration Medical Center, Aurora, Colorado, USA
| | - Mary O Whipple
- Division of General Internal Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Amy G Huebschmann
- Division of General Internal Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
- Division of Center for Women's Health Research, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Melanie Cree-Green
- Division of Center for Women's Health Research, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
- Division of Pediatric Endocrinology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Jane E B Reusch
- Division of Endocrinology, University of Colorado School of Medicine, Aurora, Colorado, USA
- Division of General Internal Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
- Division of Center for Women's Health Research, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
- Rocky Mountain Regional Veterans Administration Medical Center, Aurora, Colorado, USA
| | - Judith G Regensteiner
- Division of General Internal Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
- Division of Center for Women's Health Research, Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado, USA
| |
Collapse
|
49
|
Takeshita LY, Davidsen PK, Herbert JM, Antczak P, Hesselink MKC, Schrauwen P, Weisnagel SJ, Robbins JM, Gerszten RE, Ghosh S, Sarzynski MA, Bouchard C, Falciani F. Genomics and transcriptomics landscapes associated to changes in insulin sensitivity in response to endurance exercise training. Sci Rep 2021; 11:23314. [PMID: 34857871 PMCID: PMC8639975 DOI: 10.1038/s41598-021-98792-1] [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: 01/14/2021] [Accepted: 08/25/2021] [Indexed: 01/14/2023] Open
Abstract
Despite good adherence to supervised endurance exercise training (EET), some individuals experience no or little improvement in peripheral insulin sensitivity. The genetic and molecular mechanisms underlying this phenomenon are currently not understood. By investigating genome-wide variants associated with baseline and exercise-induced changes (∆) in insulin sensitivity index (Si) in healthy volunteers, we have identified novel candidate genes whose mouse knockouts phenotypes were consistent with a causative effect on Si. An integrative analysis of functional genomic and transcriptomic profiles suggests genetic variants have an aggregate effect on baseline Si and ∆Si, focused around cholinergic signalling, including downstream calcium and chemokine signalling. The identification of calcium regulated MEF2A transcription factor as the most statistically significant candidate driving the transcriptional signature associated to ∆Si further strengthens the relevance of calcium signalling in EET mediated Si response.
Collapse
Affiliation(s)
- Louise Y. Takeshita
- grid.10025.360000 0004 1936 8470Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown Street, Liverpool, L69 7ZB UK
| | - Peter K. Davidsen
- grid.10025.360000 0004 1936 8470Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown Street, Liverpool, L69 7ZB UK
| | - John M. Herbert
- grid.10025.360000 0004 1936 8470Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown Street, Liverpool, L69 7ZB UK
| | - Philipp Antczak
- grid.10025.360000 0004 1936 8470Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown Street, Liverpool, L69 7ZB UK ,grid.411097.a0000 0000 8852 305XCenter for Molecular Medicine Cologne, University Hospital Cologne, 50931 Cologne, Germany
| | - Matthijs K. C. Hesselink
- grid.5012.60000 0001 0481 6099Department of Nutrition and Movement Sciences, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Centre, Maastricht, The Netherlands
| | - Patrick Schrauwen
- grid.5012.60000 0001 0481 6099Department of Nutrition and Movement Sciences, NUTRIM School for Nutrition and Translational Research in Metabolism, Maastricht University Centre, Maastricht, The Netherlands
| | - S. John Weisnagel
- grid.23856.3a0000 0004 1936 8390Diabetes Research Unit, Endocrinology and Nephrology Axis, CRCHU de Québec, Université Laval, Québec City, Canada
| | - Jeremy M. Robbins
- grid.239395.70000 0000 9011 8547Division of Cardiovascular Medicine, and Cardiovascular Research Center, Beth Israel Deaconess Medical Center, Boston, MA 02215 USA
| | - Robert E. Gerszten
- grid.239395.70000 0000 9011 8547Division of Cardiovascular Medicine, and Cardiovascular Research Center, Beth Israel Deaconess Medical Center, Boston, MA 02215 USA
| | - Sujoy Ghosh
- grid.428397.30000 0004 0385 0924Centre for Computational Biology and Program in Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Mark A. Sarzynski
- grid.254567.70000 0000 9075 106XDepartment of Exercise Science, Arnold School of Public Health, University of South Carolina, Columbia, SC USA
| | - Claude Bouchard
- grid.250514.70000 0001 2159 6024Human Genomics Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA USA
| | - Francesco Falciani
- Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Crown Street, Liverpool, L69 7ZB, UK.
| |
Collapse
|
50
|
Remie CME, Janssens GE, Bilet L, van Weeghel M, Duvivier BMFM, de Wit VHW, Connell NJ, Jörgensen JA, Schomakers BV, Schrauwen-Hinderling VB, Hoeks J, Hesselink MKC, Phielix E, Houtkooper RH, Schrauwen P. Sitting less elicits metabolic responses similar to exercise and enhances insulin sensitivity in postmenopausal women. Diabetologia 2021; 64:2817-2828. [PMID: 34510226 PMCID: PMC8435176 DOI: 10.1007/s00125-021-05558-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/28/2021] [Indexed: 01/17/2023]
Abstract
AIMS/HYPOTHESIS In our current society sedentary behaviour predominates in most people and is associated with the risk of developing type 2 diabetes. It has been suggested that replacing sitting time by standing and walking could be beneficial for individuals with type 2 diabetes but the underlying mechanisms are unknown and direct comparisons with exercise are lacking. Our objective was to directly compare metabolic responses of either sitting less or exercising, relative to being sedentary. METHODS We performed a randomised, crossover intervention study in 12 overweight women who performed three well-controlled 4 day activity regimens: (1) sitting regimen (sitting 14 h/day); (2) exercise regimen (sitting 13 h/day, exercise 1 h/day); and (3) sitting less regimen (sitting 9 h/day, standing 4 h/day and walking 3 h/day). The primary outcome was insulin sensitivity measured by a two-step hyperinsulinaemic-euglycaemic clamp. We additionally performed metabolomics on muscle biopsies taken before the clamp to identify changes at the molecular level. RESULTS Replacing sitting time by standing and walking over 4 days resulted in improved peripheral insulin sensitivity, comparable with the improvement achieved by moderate-to-vigorous exercise. Specifically, we report a significant improvement in peripheral insulin sensitivity in the sitting less (~13%) and the exercise regimen (~20%), compared with the sitting regimen. Furthermore, sitting less shifted the underlying muscle metabolome towards that seen with moderate-to-vigorous exercise, compared with the sitting regimen. CONCLUSIONS/INTERPRETATIONS Replacing sitting time by standing and walking is an attractive alternative to moderate-to-vigorous exercise for improving metabolic health. TRIAL REGISTRATION ClinicalTrials.gov NCT03912922.
Collapse
Affiliation(s)
- Carlijn M E Remie
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Georges E Janssens
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Lena Bilet
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Core Facility Metabolomics, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | | | - Vera H W de Wit
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Niels J Connell
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Johanna A Jörgensen
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Bauke V Schomakers
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Core Facility Metabolomics, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Vera B Schrauwen-Hinderling
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Joris Hoeks
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Matthijs K C Hesselink
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Esther Phielix
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Riekelt H Houtkooper
- Laboratory Genetic Metabolic Diseases, Amsterdam Gastroenterology, Endocrinology, and Metabolism, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Patrick Schrauwen
- Department of Nutrition and Movement Sciences, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Maastricht, the Netherlands.
| |
Collapse
|